Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
KETO-AMIDE-BASED FIBROBLAST ACTIVATION PROTEIN-TARGETED LIGAND LINKED TO AN IMAGING OR THERAPEUTIC AGENT, COMPOSITIONS AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2024/102956
Kind Code:
A1
Abstract:
Conjugates comprising a fibroblast activation protein alpha (FAPα)-targeted ligand, a bifunctional or trifunctional linker, and an imaging or therapeutic agent; compositions comprising the conjugates; and methods of use in the imaging and treatment of FAP-positive cancer-associated fibroblasts (CAFs) and activated myofibroblasts in cancer and other fibrotic/inflammatory diseases.

Inventors:
LOW PHILIP (US)
SRINIVASARAO MADDURI (US)
MUKKAMALA RAMESH (US)
LINDEMAN SPENCER (US)
Application Number:
PCT/US2023/079298
Publication Date:
May 16, 2024
Filing Date:
November 09, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PURDUE RESEARCH FOUNDATION (US)
LOW PHILIP (US)
SRINIVASARAO MADDURI (US)
MUKKAMALA RAMESH (US)
LINDEMAN SPENCER (US)
International Classes:
A61K49/00; A61P29/00; A61P35/00; A61P43/00; C07D401/14; C07D403/14
Foreign References:
US20220001037A12022-01-06
US20220105208A12022-04-07
US20170119901A12017-05-04
US20060276435A12006-12-07
Attorney, Agent or Firm:
DEAN, Natalie, J. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS: 1. A conjugate of formula I: wherein A has the structure: wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from the group consisting of O, N, and S; R1 and R2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R5 and R6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH2=, -CH3, CH3CH2-, (CH3)2CH-, (CH3)3C-, -CH2Ph, and substituted -CH2Ph; R8-R10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO2, -SO3H, -SO2NH2, -NH2, -N3, -NH=NH, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; and R11 is selected from the group consisting of H, D, C1-C10 alkyl, C3-C10 cycloalkyl, adamantyl, substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: wherein: R12 and R16 are independently selected from the group consisting of H, D, halogen, C1-C3 alkyl, C1-C3 alkoxy, -CF3, and -C(=O)-OR23, wherein R23 is selected from the group consisting of H, D, halogen, C1-C4 alkyl, and C1-C3 alkoxy; R13, R14 and R15 are independently selected from the group consisting of H, D, halogen, -OMe, C1-C3 alkyl, C1-C3 alkoxy, -CF3, and -C(=O)-OR23, wherein R23 is selected from the group consisting of H, D, halogen, C1-C4 alkyl, and C1-C3 alkoxy; R17, R18, R20, and R21 are independently selected from H and CH3; and R19 and R22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; and B is an imaging or therapeutic agent; or is a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate thereof. 2. The conjugate of claim 1, wherein L is attached to A at any carbon atom of the functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, and is or comprises a 1° amine, a 2° amine, a functionalized alkyl, or a functionalized cycloalkyl. 3. The conjugate of claim 1, wherein A has the formula IV or V: wherein: R2 is selected from the group consisting of -H, -D, -OH, -F, -Cl, -Br, I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R5 and R6 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH2=, -CH3, CH3CH2-, (CH3)2CH- , and (CH3)3C-; R11 is selected from the group consisting of

R23-R26 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -CF3, -NO2, -SO3H, -SO2NH2, -NH2, -N3, -NH=NH-, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl. 4. The conjugate of claim 3, wherein L is attached to A at any carbon atom of the functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, and is or comprises a 1° amine, a 2° amine, a functionalized alkyl, or a functionalized cycloalkyl. 5. The conjugate of any one of claims 1-4, wherein B comprises a radical of a transforming growth factor beta (TGF ^)/Smad inhibitor, a Wnt/ ^-catenin inhibitor, a vascular endothelial growth factor receptor 1 (VEGFR1) inhibitor, a VEGFR2 inhibitor, a vascular endothelial growth factor receptor 3 (VEGFR3) inhibitor, a fibroblast growth factor receptor 1 (FGFR1) inhibitor, a fibroblast growth factor receptor 2 (FGFR2) inhibitor, a platelet-derived growth factor receptor (PDGFR) inhibitor, a FAK inhibitor, a rho kinases (ROCK) inhibitor, a PDGFR inhibitor, a toll-like receptor (TLR) agonist, an NF-κB (nuclear factor kappa-light- chain-enhancer of activated B cells) inhibitor, an inhibitor of collagen synthesis, or an angiogenesis inhibitor. 6. The conjugate of any one of claims 1-4, wherein B comprises a chemotherapeutic agent, an anti-fibrotic agent, an anti-cancer agent, or an immunotherapeutic agent. 7. The conjugate of any one of claims 1-4, wherein: B comprises a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope, wherein said metal or isotope is suitable for radio-imaging, positron emission tomography (PET) imaging, single-photon emission computerized tomography (SPECT) imaging, radiotherapy, or magnetic resonance imaging, and/or comprises a fluorescent imaging agent, a photodynamic imaging agent, an optical imaging agent, a photosensitizer, or a radiosensitizer. 8. The conjugate of claim 1, wherein B comprises a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope, and comprises a fluorescent imaging agent, a photodynamic imaging agent, an optical imaging agent, a photosensitizer, or a radiosensitizer. 9. The conjugate of claim 8, wherein the fluorescent imaging agent is selected from the group consisting of carbocyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine and merocyanine, polymethine, coumarine, rhodamine, xanthene, fluorescein, borondipyrromethane (BODIPY), Indocyanine green (ICG), CyS, CyS.S, Cy7, VivoTag-680, VivoTag-S680, VivoTag-S7S0, AlexaFluor660, AlexaFluor680, AlexaFluor700, AlexaFluor7S0, 10 AlexaFluor790, Dy677, Dy676, Dy682, Dy7S2, Dy780, DyLightS47, Dylight647, HiLyte Fluor 647, HiLyte Fluor 680, HiLyte Fluor 7S0, IRDye 800CW, IRDye 800RS, IRDye 700DX, ADS780WS, ADS830WS, ADS832WS and S0456. 10. The conjugate of claim 8, wherein the fluorescent imaging agent has a structure selected from:

11. The conjugate of claim 8, wherein the fluorescent imaging agent has a structure selected from: , and

12. The conjugate of claim 8, wherein the photosensitizer has a structure selected from:

13. The conjugate of claim 8, wherein the metal chelating group is selected from the group consisting of DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) or a derivative thereof; TETA (1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid) or a derivative thereof; SarAr (1-N-(4-Aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]- eicosane-1,8-diamine or a derivative thereof; NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) or a derivative thereof; NETA (4-[2-(bis-carboxymethylamino)-ethyl]-7-carboxymethyl- [1,4,7]triazonan-1-yl) acetic acid or a derivative thereof TRAP (1,4,7-triazacyclononane-1,4,7- tris[methyl(2-carboxyethyl)phosphinic acid) or a derivative thereof; HBED (N,N0-bis(2- hydroxybenzyl)-ethylenediamine-N,N0-diacetic acid) or a derivative thereof; 2,3-HOPO (3- hydroxypyridin-2-one) or a derivative thereof; PCTA (3,6,9,15-tetraazabicyclo[9.3.1]- pentadeca-1(15),11,13-triene-3,6,9,-triacetic acid) or a derivative thereof; DFO (desferrioxamine) or a derivative thereof; DTPA (diethylenetriaminepentaacetic acid) or a derivative thereof; OCTAPA (N,N0-bis(6-carboxy-2-pyridylmethyl)-ethylenediamine-N,N0- diacetic acid) or a derivative thereof; H2-MACROPA (N,N'-bis[(6-carboxy-2-pyridipmethyl]- 4,13-diaza-18-crown-6) or a derivative thereof; S-2-(4-Isothiocyanatobenzyl)-1,4,7,10- tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA) or a derivative thereof; H2dedpa (1,2-[[carboxy)-pyridin-2-yl]-methylamino]ethane or a derivative thereof; and EC20-head comprising β-l-diaminopropionic acid, aspartic acid, and cysteine. 14. The conjugate of claim 8, wherein the metal chelating group is bound to 11C, 13C, 13N, 15O, 18F, 32P, 44Sc, 47Sc, 52Mn, 55Co, 60Co, 64Cu, 67Cu, 67Ga, 68Ga, 86Y, 89Sr, 89Zr, 90Y, 99mTc, 111In, 114mIn, 117mSn, 123I, 124I, 125I, 131I, 149Tb, 153Sm, 152Tb, 155Tb, 161Tb, 169Er, 177Lu, 186Re, 188Re, 211At, 212Pb, 212Bi, 213Bi, 223Ra, 224Ra, 225Ab, 225Ac, or 227Th. 15. The conjugate of claim 8, wherein B comprises a structure selected from the group consisting of

16. The conjugate of claim 8, wherein said metal chelating group has a structure selected from:

17. The conjugate of claim 6, wherein said anti-cancer agent is selected from the group consisting of

18. The conjugate of claim 8, wherein the radiosensitizer is selected from:

19. The conjugate of claim 8, wherein the radiosensitizer is selected from the group consisting of topoisomerase inhibitor (e.g., camptothecin, topotecan), hypoxia-activated anthraquinone AQ4N, an alkylating agent (e.g., temozolomide), a drug affecting a DNA repair pathway (e.g., poly(ADP ribose)polymerase inhibitor or AG14,361), and a PRMT5 inhibitor (e.g., JNJ-64619178). 20. The conjugate of claim 6, wherein the chemotherapeutic agent is selected from the group consisting of 21. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula: wherein n is an integer from 0 to 10.

22. The conjugate of any one of claims 1-4 and 8-20, where L is or comprises a moiety of the formula: wherein n is an integer from 0 to 10. 23. The conjugate of any one of claims 1-4, wherein L is or comprises a moiety of the formula: . 24. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula: wherein n is an integer from 0 to 10.

25. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula: . 26. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula:

27. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula: wherein n is an integer from 0 to 20. 28. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises a moiety of the formula: , wherein n is an integer from 0 to 20.

29. The conjugate of any one of claims 1-4 and 8-20, wherein L is a linker that can be cleaved. 30. The conjugate of claim 27, wherein L can be cleaved reductively, oxidatively, or enzymatically. 31. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises an oxime ester. 32. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises a hydrazone. 33. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises polyethylene glycoln (PEGn), wherein n = 0-36. 34. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises a peptide, a peptidoglycan, an alkyl, or a sugar. 35. The conjugate of any one of claims 1-4 and 8-20, wherein L is or comprises:

36. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises a structure selected from: wherein: R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. 37. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises the structure: wherein: R31 is H or C1-C6 alkyl; and R29a, R29b, R30a, and R30b are independently selected from the group consisting of H and C1-C6 alkyl.

38. The conjugate of any one of claims 1-4 and 8-20, wherein L comprises a structure selected from: wherein: R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. 39. The conjugate of claim 1, which has the formula (II): , wherein C is a pharmacokinetic extender. 40. The conjugate of claim 39, wherein C is an albumin binder, a plasma protein binder, or a hapten. 41. The conjugate of claim 39, wherein the albumin binder is or comprises albumin binding domain 035 (ABD035), albumin binding domain Con (ABDCon), a designed ankyrin repeat protein (DARPin), a disulfide stabilized Fv fragment (dsFv), an anti-albumin antibody CA645, an anti-human serum albumin nanobody, or variable new antigen receptor E06 (VNAR E06). 42. A pharmacokinetically extended conjugate comprising the conjugate of any one of claims 1-38 and which has the formula (II): , wherein: A has the structure: wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from the group consisting of O, N, and S; R1 and R2 are independently selected from the group consisting of -H, -D - OH, - F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, - Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R5 and R6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH2=, -CH3, CH3CH2-, (CH3)2CH-, (CH3)3C-, -CH2Ph, and substituted -CH2Ph; R8-R10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO2, -SO3H, -SO2NH2, -NH2, -N3, -NH=NH, -C1-6 alkyl, -O-C1-6 alkyl, and -S-C1-6 alkyl; and R11 is selected from the group consisting of H, D, Cl-Cl0 alkyl, C3-Cl0 cycloalkyl, adamantyl, substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: wherein: R12 and R16 are independently selected from the group consisting of H, D, halogen, C1-C3 alkyl, C1-C3 alkoxy, -CF3, and -C(=O)-OR23, wherein R23 is selected from the group consisting of H, D, halogen, C1-C4 alkyl, and C1-C3 alkoxy; R13, R14 and R15 are independently selected from the group consisting of H, D, halogen, -OMe, C1-C3 alkyl, C1-C3 alkoxy, -CF3, and -C(=O)-OR23, wherein R23 is selected from the group consisting of H, D, halogen, C1-C4 alkyl, and C1-C3 alkoxy; R17, R18, R20, and R21 are independently selected from H and CH3; and R19 and R22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; B is an imaging or therapeutic agent; and C is a pharmacokinetic extender; or is a stereoisomer or a pharmaceutically acceptable salt, solvate, or hydrate thereof. 43. The pharmacokinetically extended conjugate of claim 42, wherein C is an albumin binder, a plasma protein binder, or a hapten. 44. The pharmacokinetically extended conjugate of claim 42, wherein the albumin binder is or comprises albumin binding domain 035 (ABD035), albumin binding domain Con (ABDCon), a designed ankyrin repeat protein (DARPin), a disulfide stabilized Fv fragment (dsFv), an anti-albumin antibody CA645, an anti-human serum albumin nanobody, or a variable new antigen receptor E06 (VNAR E06).

45. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises: . 46. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises:

wherein: each of R12-19 is independently -H, -C1-C6 alkyl, -F, -Cl, -Br, -I, -CN, -CHO, -B(OH)2, -C(O)alkyl, -C(O)aryl-, -C=C-C(O)aryl, -C=C-S(O)2aryl, -CO2H, -SO3H, -SO2NH2, -PO3H2, or -SO2F; and each of R20 and R21 is independently -H, -C1-C6 alkyl, -F, -Cl, -Br, -I, -O-C1-6 alkyl, -CN, -CHO, -B(OH)2, -C=C-C(O)aryl, -C=C-S(O)2aryl, -CO2H, -SO3H, -SO2NH2, -PO3H2, -SO2F, O CF3, or 0-14. 47. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises: .

48. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises: 49. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises: 50. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C comprises a radical of polyethylene glycoln (PEGn), wherein n = 0-32, a peptide, a petidoglycan, or a saccharide. 51. The pharmacokinetically extended conjugate of any one of claims 39-41, wherein C is or comprises:

52. The pharmacokinetically extended conjugate of any one of claims 42-44, wherein C is or comprises . 53. The pharmacokinetically extended conjugate of claim 42, wherein the hapten is recognized by an autologous antibody. 54. The pharmacokinetically extended conjugate of claim 42, wherein the hapten is selected from the group consisting of rhamnose, an α-galactosyl moiety, a dinitrophenyl (DNP) moiety, and a trinitrophenyl (TNP) moiety. 55. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: O H O O O H N N O O n n , wherein n is an integer from 0 to 10. 56. The conjugate of claim 42, where L is or comprises a moiety of the formula: wherein n is an integer from 0 to 10. 57. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: .

58. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: wherein n is an integer from 0 to 10. 59. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: . 60. The conjugate of claim 42, wherein L is or comprises a moiety of the formula:

61. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: wherein n is an integer from 0 to 20. 62. The conjugate of claim 42, wherein L is or comprises a moiety of the formula: O wherein n is an integer from 0 to 20. 63. The conjugate of claim 42, wherein L is a linker that can be cleaved. 64. The conjugate of claim 63, wherein L can be cleaved reductively, oxidatively, or enzymatically. 65. The conjugate of claim 42, wherein L comprises an oxime ester. 66. The conjugate of claim 42, wherein L comprises a hydrazone. 67. The conjugate of claim 42, wherein L comprises polyethylene glycoln (PEGn), wherein n = 1-36. 68. The conjugate of claim 42, wherein L comprises a peptide, a peptidoglycan, an alkyl, or a sugar.

69. The conjugate of claim 42, wherein L is or comprises: . 70. The conjugate of claim 42, wherein L comprises a structure selected from the group consisting of , wherein: R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. 71. The conjugate of claim 42, wherein L comprises the structure: wherein: R31 is H or C1-C6 alkyl; and R29a, R29b, R30a, and R30b are independently selected from the group consisting of H and C1-C6 alkyl. 72. A conjugate having a structure selected from:

73. A conjugate having a structure of , wherein n = 1-5.

74. A conjugate having a structure selected from:

wherein n = 1-5.

75. A conjugate having a structure selected from:

wherein n = 1-5.

76. A conjugate having a structure selected from: .

77. A conjugate having a structure selected from: or , or

r

or , or wherein m = 1-10 and p = 1-20. 78. A conjugate having a structure selected from: or

, wherein m = 1-10 and p = 1-20.

79. A conjugate having a structure of: M

80. A conjugate having a structure of:

81. A conjugate having a structure selected from: ,

,

,

, ,

,

, wherein: n = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. 82. A conjugate having a structure selected from:

wherein: p = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. 83. A conjugate having a structure selected from: , wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging.

84. A conjugate having a structure selected from: O M wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. 85. A conjugate having a structure selected from: ,

,

, wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. 86. A conjugate having a structure selected from: , wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. 87. A conjugate having a structure selected from: .

88. A conjugate having a structure selected from: . 89. A conjugate having a structure selected from: .

90. A conjugate having a structure selected from: . 91. A conjugate having a structure selected from: .

92. A conjugate having a structure selected from: , , wherein n = 0-20. 93. A pharmaceutical composition comprising a conjugate of any one of claims 1-92 or a pharmaceutically acceptable salt, solvate, hydrate, or stereoisomer thereof, and a pharmaceutically acceptable carrier. 94. A method of imaging cells displaying fibroblast activation protein (FAP) on their surfaces (FAP+ cells) in a subject comprising: administering to the subject (i) a conjugate of any one of claims 1-4, 8-20, 39-41, and 72- 92, (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, or (iii) a pharmaceutical composition comprising a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92 and carrier; and imaging the conjugate or stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate thereof bound to FAP on surfaces of cells displaying FAP. 95. The method of claim 94, wherein imaging is performed by magnetic resonance imaging (MRI), ultrasound, X-ray, optical imaging, Computed Tomography (CT), Single Photon Emission Computed Tomography (SPECT), Positron Emission Tomography (PET), Fluorescence Resonance Energy Transfer (FRET), or any combination of two or more of the foregoing. 96. The method of claim 94 or 95, further comprising assessing or monitoring efficacy of a treatment administered to the subject. 97. The method of claim 94, further comprising administering radiotherapy to the subject, wherein the radiotherapy can be administered before, concurrent with, or sequential to administering the conjugate, stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of the conjugate, or the composition to the subject. 98. A method of treating cancer in a subject comprising administering to the subject an effective amount of: (i) a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, or (iii) a pharmaceutical composition comprising a conjugate of any one of claims 1-4, 8- 20, 39-41, and 72-92 and carrier; whereupon the subject is treated for cancer. 99. The method of claim 98, wherein the cancer is glioblastoma. 100. The method of claim 98, wherein the cancer is colorectal cancer. 101. The method of claim 98, wherein the cancer is breast cancer. 102. The method of claim 98, further comprising imaging the conjugate or stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate thereof bound to FAP on surfaces of cells displaying FAP.

103. The method of claim 98, further comprising administering an effective amount of a second anti-cancer therapy to the subject, wherein the second anti-cancer therapy comprises one or more of radiotherapy, brachytherapy, photodynamic therapy, photothermal therapy, focal ablation therapy including cryoablation, focal laser ablation and high-frequency ultrasound ablation, chemotherapy, and immunotherapy. 104. The method of claim 98, wherein B of the conjugate comprises a chemotherapeutic agent, an anti-cancer agent, or an immunotherapeutic agent. 105. A method of treating fibrosis in a subject comprising administering to the subject an effective amount of: (i) a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, or (iii) a pharmaceutical composition comprising a conjugate of any one of claims 1-4, 8- 20, 39-41, and 72-92 and carrier; whereupon the subject is treated for fibrosis. 106. A method of treating an inflammatory disease or disorder in a subject comprising administering to the subject an effective amount of: (i) a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92, or (iii) a pharmaceutical composition comprising a conjugate of any one of claims 1-4, 8- 20, 39-41, and 72-92 and carrier; whereupon the subject is treated for an inflammatory disease or disorder. 107. Use of (i) a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92; (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92; or (iii) a pharmaceutical composition comprising a conjugate of any one of claims 1-4, 8-20, 39-41, and 72-92 and carrier in the manufacture of a medicament for the treatment of cancer, fibrosis, or an inflammatory disease or disorder in a subject.

108. The use of claim 107, wherein the cancer is glioblastoma, breast cancer, or colorectal cancer. 109. A conjugate having a structure of: , wherein n = 1-20.

Description:
KETO-AMIDE-BASED FIBROBLAST ACTIVATION PROTEIN-TARGETED LIGAND LINKED TO AN IMAGING OR THERAPEUTIC AGENT, COMPOSITIONS AND METHODS OF USE PRIORITY [0001] This patent application is related to and claims the priority benefit of U.S. Provisional Patent Application No. 63/423,905 filed November 9, 2022. The content of the foregoing application is hereby incorporated by reference in its entirety into this disclosure. TECHNICAL FIELD [0002] The present disclosure relates to conjugates comprising a fibroblast activation protein α (FAP-α)-targeted ligand, a bifunctional or trifunctional linker, and an imaging or therapeutic agent, compositions comprising the conjugates, and methods of using the conjugates/compositions to image and/or treat FAP-positive cancer-associated fibroblasts (CAFs) and activated myofibroblasts in cancer and other fibrotic/inflammatory diseases. BACKGROUND [0003] The survival and proliferation of tumor is dependent on the percentage of tumor stroma (TSP). A high TSP is associated with poorer, long-term patient survival compared to low TSP (> 50% vs. ^ 50% respectively). The TSP is also a significant prognostic factor for tumor relapse, growth, and metastasis. [0004] Cancer-associated fibroblasts (CAFs) are abundant in the tumor stroma and perform several important functions to promote tumorigenesis. These functions include cytokine secretion and extracellular matrix (ECM) production and remodeling. Resulting angiogenesis promotes tumor growth, signaling factors increase chemoresistance, increased ECM density creates an immunosuppressive environment, and enhanced cell motility directs metastasis. These mechanisms are well-documented and parallel the behavior of pathogenic fibroblasts in fibrotic diseases. [0005] One of the most prevalent markers of CAFs is fibroblast activation protein alpha (FAPα). FAP ^ is a serine protease primarily found on the cell surface of activated fibroblasts in diseases such as fibrosis, rheumatoid arthritis, wound healing, and cancer. More than 90% of epithelial carcinomas show FAP ^ expression in immunohistochemical (IHC) staining. Additional FAP ^ expression has been found in a subset of primary glioma cell cultures and tumor-associated macrophages (TAMs). However, FAP ^ expression is very low or nonexistent in the majority of adult tissues. Therefore, because the expression is restricted to the surface of diseased cells, such as cancerous cells, FAPα is uniquely qualified as a receptor for selectively delivering pharmacotherapeutics and imaging agents to tumors via ligand-targeting. [0006] FAPα is already being exploited as a ligand-target for imaging with near-infrared (NIR) dyes and positron emission tomography (PET) agents. Similarly, therapies to kill tumor cells, such as antimitotic or radiotherapeutic agents, are being delivered to the tumor microenvironment by targeting FAPα. The ligands currently in use, however, have low tumor retention, poor signal to background ratio, and/or unwanted uptake in healthy tissues. [0007] In view of the above, it is an object to provide a conjugate comprising a FAPα-targeted ligand linked to an imaging or therapeutic agent. The conjugates hereof offer increased tumor retention and better signal to background ratio. This and other objects and advantages, as well as inventive features, will be apparent from the detailed description provided herein. SUMMARY [0008] Provided are conjugates of formula I: wherein A has the structure: wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from the group consisting of O, N, and S; R 1 and R 2 are independently selected from the group consisting of -H, -D, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R5 and R6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of -H, -D, OH, CH 2 =, -CH 3 ,CH 3 CH 2 -, (CH 3 )2CH- , (CH 3 )3C-, -CH 2 Ph, and substituted -CH 2 Ph; R 8 -R 10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R11 is selected from the group consisting of -H, -D, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, adamantyl substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: wherein: R12 and R16 are independently selected from the group consisting of -H, -D, halogen, Cl- C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 13 , R 14 and R 15 are independently selected from the group consisting of -H, -D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-O R 23 , wherein R 23 is selected from the group consisting of -H, -D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from -H and -CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; and B is an imaging or therapeutic agent; or a stereoisomer or a pharmaceutically acceptable salt thereof. [0009] L can be attached to A at any carbon atom of the functionalized 5- to 10-membered N- containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, a 1° amine, a 2° amine, a functionalized alkyl, or a functionalized cycloalkyl. A can have the formula IV or V:

wherein: R 2 is selected from the group consisting of -H, -D, -OH, -F, -Cl, -Br, I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 3 and R 4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, - I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R5 and R6 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, - I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 )2CH- , and (CH 3 ) 3 C-; R11 is selected from the group consisting of

R 23 -R26 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -CF 3 , -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH-, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1- 6 alkyl. [0010] B can comprise a radical of a transforming growth factor beta (TGF ^)/Smad inhibitor, a Wnt/ ^-catenin inhibitor, a vascular endothelial growth factor receptor 1 (VEGFR1) inhibitor, a VEGFR2 inhibitor, a vascular endothelial growth factor receptor 3 (VEGFR3) inhibitor, a fibroblast growth factor receptor 1 (FGFR1) inhibitor, a fibroblast growth factor receptor 2 (FGFR2) inhibitor, a platelet-derived growth factor receptor (PDGFR) inhibitor, a FAK inhibitor, a rho kinases (ROCK) inhibitor, a PDGFR inhibitor, a toll-like receptor (TLR) agonist, an NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) inhibitor, an inhibitor of collagen synthesis, or an angiogenesis inhibitor. [0011] B can comprise a chemotherapeutic agent, an anti-fibrotic agent, an anti-cancer agent, or an immunotherapeutic agent. B can comprise a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope, wherein said metal or isotope is suitable for radio- imaging, positron emission tomography (PET) imaging, single-photon emission computerized tomography (SPECT) imaging, radiotherapy, or magnetic resonance imaging, a fluorescent imaging agent, a photodynamic imaging agent, an optical imaging agent, a photosensitizer, or a radiosensitizer. In certain embodiments, B comprises a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope, and comprises a fluorescent imaging agent, a photodynamic imaging agent, an optical imaging agent, a photosensitizer, or a radiosensitizer. [0012] The fluorescent imaging agent can be selected from the group consisting of carbocyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine and merocyanine, polymethine, coumarine, rhodamine, xanthene, fluorescein, borondipyrromethane (BODIPY), Indocyanine green (ICG), CyS, CyS.S, Cy7, VivoTag-680, VivoTag-S680, VivoTag-S7S0, AlexaFluor660, AlexaFluor680, AlexaFluor700, AlexaFluor7S0, 10 AlexaFluor790, Dy677, Dy676, Dy682, Dy7S2, Dy780, DyLightS47, Dylight647, HiLyte Fluor 647, HiLyte Fluor 680, HiLyte Fluor 7S0, IRDye 800CW, IRDye 800RS, IRDye 700DX, ADS780WS, ADS830WS, ADS832WS and S0456. The fluorescent imaging agent can have a structure selected from:

[0013] The fluorescent imaging agent can have a structure selected from:

[0014] The photosensitizer can have a structure selected from:

[0015] The metal chelating group can be selected from the group consisting of DOTA (1,4,7,10- tetraazacyclododecane-1,4,7,10-tetraacetic acid) or a derivative thereof; TETA (1,4,8,11- tetraazacyclotetradecane-1,4,8,11-tetraacetic acid) or a derivative thereof; SarAr (1-N-(4- Aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]-eicosane- 1,8-diamine or a derivative thereof; NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) or a derivative thereof; NETA (4- [2-(bis-carboxymethylamino)-ethyl]-7-carboxymethyl-[1,4,7]tr iazonan-1-yl) acetic acid or a derivative thereof TRAP (1,4,7-triazacyclononane-1,4,7-tris[methyl(2-carboxyethyl)ph osphinic acid) or a derivative thereof; HBED (N,N0-bis(2-hydroxybenzyl)-ethylenediamine-N,N0-diacetic acid) or a derivative thereof; 2,3-HOPO (3-hydroxypyridin-2-one) or a derivative thereof; PCTA (3,6,9,15-tetraazabicyclo[9.3.1]-pentadeca-1(15),11,13-trien e-3,6,9,-triacetic acid) or a derivative thereof; DFO (desferrioxamine) or a derivative thereof; DTPA (diethylenetriaminepentaacetic acid) or a derivative thereof; OCTAPA (N,N0-bis(6-carboxy-2-pyridylmethyl)-ethylenediamine- N,N0-diacetic acid) or a derivative thereof; H2-MACROPA (N,N'-bis[(6-carboxy-2- pyridipmethyl]-4,13-diaza-18-crown-6) or a derivative thereof; S-2-(4-Isothiocyanatobenzyl)- 1,4,7,10-tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA) or a derivative thereof; H2dedpa (1,2-[[carboxy)-pyridin-2-yl]-methylamino]ethane or a derivative thereof; and EC20- head comprising β-l-diaminopropionic acid, aspartic acid, and cysteine. The metal chelating group can be bound to 11 C, 13 C, 13 N, 15 O, 18 F, 32 P, 44 Sc, 47 Sc, 52 Mn, 55 Co, 60 Co, 64 Cu, 6 7Cu, 67 Ga, 6 8 Ga, 86 Y, 89 Sr, 89 Zr, 90 Y, 99m Tc, 111 In, 114m In, 117m Sn, 123 I, 124 I, 125 I, 131 I, 149 Tb, 153 Sm, 152 Tb, 155 Tb, 1 61 Tb, 169 Er, 177 Lu, 186 Re, 188 Re, 211 At, 212 Pb, 212 Bi, 213 Bi, 223 Ra, 224 Ra, 225 Ab, 225 Ac, or 227 Th. B can comprise a structure selected from the group consisting of [0016] The metal chelating group can have a structure selected from:

[0017] In certain embodiments, B comprises a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope comprising a radiosensitizer. The radiosensitizer can be selected from:

[0018] The radiosensitizer can be selected from a topoisomerase inhibitor (e.g., camptothecin, topotecan), hypoxia-activated anthraquinone AQ4N, an alkylating agent (e.g., temozolomide), a drug affecting a DNA repair pathway (e.g., poly(ADP ribose)polymerase inhibitor, AG14,361), and a PRMT5 inhibitor (e.g., JNJ-64619178). [0019] In certain embodiments, B comprises a chemotherapeutic agent. The chemotherapeutic agent can be selected from:

[0020] B can comprise an anti-cancer agent. The anti-cancer agent can be effective against cancer cells, cancer-associated fibroblasts, or factors in the tumor microenvironment. The anti-cancer agent can be selected from:

wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: . [0022] L can be or can comprise a moiety of any one of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula:

[0023] L can be or can comprise a moiety of the formula: [0024] L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 20. [0025] L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 20. L can be a linker that can be cleaved. L can be cleaved reductively, oxidatively, or enzymatically. L can comprise an oxime ester. L can comprise a hydrazone. L can comprise polyethylene glycoln (PEGn), wherein n = 0-36. L can comprise a peptide, a peptidoglycan, an alkyl, or a sugar. L can be or can comprise: [0026] L can be or can comprise: wherein: R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. [0027] L can comprise the structure: wherein: R 31 is H or C 1 -C 6 alkyl; and R 29a , R 29b , R 30a, and R 30b are independently selected from the group consisting of H and C1-C6 alkyl. [0028] L can comprise a structure selected from: whe R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. [0029] In certain embodiments, the conjugate can further comprise the formula (II): wherein C is a pharmacokinetic extender. C can be an albumin binder, a plasma protein binder, or a hapten. The albumin binder can be or comprise albumin binding domain 035 (ABD035), albumin binding domain Con (ABDCon), a designed ankyrin repeat protein (DARPin), a disulfide stabilized Fv fragment (dsFv), an anti-albumin antibody CA645, an anti-human serum albumin nanobody, or variable new antigen receptor E06 (VNAR E06). [0030] Also provided is a pharmacokinetically extended conjugate which has the formula (II): wherein: A has the structure: wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from the group consisting of O, N, and S; R1 and R 2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, - Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R5 and R6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 ) 2 CH-, (CH 3 ) 3 C-, -CH 2 Ph, and substituted -CH 2 Ph; R8-R10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R11 is selected from the group consisting of H, D, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, adamant substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: wherein: R12 and R16 are independently selected from the group consisting of H, D, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R13, R14 and R15 are independently selected from the group consisting of H, D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from H and CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; B is an imaging or therapeutic agent; and C is a pharmacokinetic extender; or is a stereoisomer or a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0031] A, B, and L of formula (II) can be or comprise any A, B, or L, respectively, described herein in connection with formula (I). C can be an albumin binder, a plasma protein binder, or a hapten. The albumin binder is or comprises ABD035, ABDCon, a DARPin, a dsFv, an anti- albumin antibody CA645, an anti-human serum albumin nanobody, or a VNAR E06. C can be or can comprise any of the following structures:

[0032] C can be or can comprise:

wherein: each of R12-19 is independently -H, -C1-C6 alkyl, -F, -Cl, -Br, -I, -CN, -CHO, -B(OH)2, -C(O)alkyl, -C(O)aryl-, -C=C-C(O)aryl, -C=C-S(O)2aryl, -CO2H, -SO 3 H, -SO 2 NH 2 , -PO3H2, or -SO 2 F; and each of R 20 and R 21 is independently -H, -C1-C6 alkyl, -F, -Cl, -Br, -I, -O-C 1-6 alkyl, -CN, -CHO, -B(OH) 2 , -C=C-C(O)aryl, -C=C-S(O) 2 aryl, -CO 2 H, -SO 3 H, -SO 2 NH 2 , -PO 3 H 2 , -SO 2 F, [0033] C can be or can comprise: [0034] C can be or can comprise: [0035] C can comprise a radical o a peptide, a petidoglycan, or a saccharide. C can be or can comprise: . C can be or can comprise: . The hapten can be recognized by an autologous antibody. The hapten can be selected from the group consisting of rhamnose, an α-galactosyl moiety, a dinitrophenyl (DNP) moiety, and a trinitrophenyl (TNP) moiety. [0036] L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: [0037] L can be or can comprise a moiety of the formula:

wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: . L can be or can comprise a moiety of the formula: [0038] L can be or can comprise a moiety of the formula:

wherein n is an integer from 0 to 20. L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 20. L can be a linker that can be cleaved. L can be cleaved reductively, oxidatively, or enzymatically, L can comprise an oxime ester. L can comprise hydrazone. L can comprise (PEG)n, wherein n = 1-36. L can comprise a peptide, a peptidoglycan, an alkyl, or a sugar. L can be or can comprise: [0039] L can comprise a structure selected from: wherein: R 27 and R 28 are independently selected from the group consisting of H and C 1 -C 6 alkyl; and Z is an integer from 1 to 8. L can comprise the structure: wherein: R31 is H or C1-C6 alkyl; and R29a, R29b, R30a, and R30b are independently selected from the group consisting of H and C1-C6 alkyl.

[0040] Still further provided is a conjugate having a structure selected from:

[0041] Still further provided is a conjugate having a structure of:

wherein n = 1-5. [0042] Still further provided is a conjugate having a structure selected from:

[0043] Still further provided is a conjugate having a structure selected from:

[0044] Still further provided is a conjugate having a structure selected from: [0045] Still further provided is a conjugate having a structure selected from:

wherein m = 1-10 and p = 1-20. [0046] Still further provided is a conjugate having a structure selected from:

, wherein m = 1-10 and p = 1-20. [0047] Still further provided is a conjugate having a structure of:

[0048] Still further provided is a conjugate having a structure of:

[0049] Still further provided is a conjugate having a structure selected from:

wherein: n = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance inmaging.

[0050] In certain embodiments, the conjugate has a structure selected from: wherein: p = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0051] In certain embodiments, the conjugate has a structure selected from: wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0052] In certain embodiments, the conjugate has a structure selected from: wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0053] In certain embodiments, the conjugate has a structure selected from:

, ,

, or wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0054] In certain embodiments, the conjugate has a structure selected from:

wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0055] In certain embodiments, the conjugate has a structure selected from:

[0056] In certain embodiments, the conjugate has a structure selected from: [0057] In certain embodiments, the conjugate has a structure selected from:

[0058] In certain embodiments, the conjugate has a structure selected from: [0059] In certain embodiments, the conjugate has a structure selected from:

[0060] Still further are conjugates having a structure selected from:

wherein n = 0-20. [0061] Still further provided is a pharmaceutical composition comprising a conjugate described herein or a pharmaceutically acceptable salt, solvate, hydrate, or stereoisomer thereof, and a pharmaceutically acceptable carrier. [0062] In view of the above, a method of imaging cells displaying fibroblast activation protein (FAP) on their surfaces (FAP+ cells) in a subject (e.g., in need of imaging) is provided. The method can comprise administering to the subject (i) a conjugate described herein, (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate described herein, or (iii) a pharmaceutical composition comprising a conjugate described herein and carrier (e.g., a pharmaceutically acceptable carrier); and imaging the conjugate or stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate thereof bound to FAP on surfaces of cells displaying FAP. Imaging can be performed by magnetic resonance imaging (MRI), ultrasound, X-ray, optical imaging, Computed Tomography (CT), Single Photon Emission Computed Tomography (SPECT), Positron Emission Tomography (PET), Fluorescence Resonance Energy Transfer (FRET), or any combination of two or more of the foregoing. The method of imaging can further comprise assessing or monitoring efficacy of a treatment administered to the subject. In certain embodiments, the method comprises administering radiotherapy to the subject, wherein the radiotherapy can be administered before, concurrent with, or sequential to administering the conjugate, stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of the conjugate, or the composition to the subject. [0063] Also in view of the above, several methods of treatment are provided. In an embodiment, a method of treating cancer in a subject is provided. In another embodiment, a method of treating fibrosis in a subject is provided. In yet another embodiment, a method of treating an inflammatory disease or disorder in a subject is provided. The method can comprise administering to the subject an effective amount of a conjugate described herein, a stereoisomer or a pharmaceutically acceptable salt, solvate, or hydrate of a conjugate hereof, or a pharmaceutical composition comprising same and a carrier; whereupon the subject is treated for cancer. B of the conjugate can comprise, for example, a chemotherapeutic agent, an anti-cancer agent, or an immunotherapeutic agent, or any other active agent that may be useful for delivery to a targeted site (e.g., a tumor microenvironment, a tumor, etc.). [0064] The cancer can be a glioblastoma. The cancer can be a colorectal cancer. The cancer can be breast cancer. In certain embodiments, the method of treatment can further comprise imaging the conjugate or stereoisomer or pharmaceutically acceptable salt, hydrate or solvate thereof bound to FAP on surfaces of cells displaying FAP (e.g., cells of the subject). In certain embodiments, the method can further comprise administering an effective amount of a second anti-cancer therapy to the subject, wherein the second anti-cancer therapy comprises one or more of radiotherapy, brachytherapy, photodynamic therapy, photothermal therapy, focal ablation therapy including cryoablation, focal laser ablation and high-frequency ultrasound ablation, chemotherapy, and immunotherapy. [0065] A method of treating fibrosis in a subject is also provided. Such method of treatment can comprise administering to the subject an effective amount of: (i) a conjugate hereof; (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof; or (iii) a pharmaceutical composition comprising a conjugate hereof or a stereoisomer, or pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof and a carrier, whereupon the subject is treated for fibrosis. [0066] A method of treating an inflammatory disease or disorder in a subject is also provided. In certain embodiments, such method comprises administering to the subject an effective amount of: (i) a conjugate hereof; (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof; or (iii) a pharmaceutical composition comprising a conjugate hereof or a stereoisomer, or pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof and a carrier, whereupon the subject is treated for an inflammatory disease or disorder. [0067] Uses of (i) a conjugate hereof; (ii) a stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof; or (iii) a pharmaceutical composition comprising a conjugate hereof or a stereoisomer, or pharmaceutically acceptable salt, hydrate, or solvate of a conjugate hereof, and carrier in the manufacture of a medicament for the treatment of cancer, fibrosis, or an inflammatory disease or disorder in a subject are also provided. The cancer can be glioblastoma, breast cancer, or colorectal cancer, for example. BRIEF DESCRIPTION OF DRAWINGS [0068] The disclosed embodiments and other features, advantages, and aspects contained herein, and the matter of attaining them, will become apparent in light of the following detailed description of various exemplary embodiments of the present disclosure. Such detailed description will be better understood when taken in conjunction with the accompanying drawings. [0069] Fig.1 is a general scheme for the synthesis of Conjugates 2 and 3 hereof. [0070] Fig.2 shows a general scheme for the synthesis of Conjugates 5 and 6 hereof. [0071] Fig.3 shows the secondary and three-dimensional (3D) structure of Conjugate 2. [0072] Fig.4 shows the secondary and 3D structure of Conjugate 3. [0073] Fig.5 shows the secondary and 3D structure of Conjugate 4. [0074] Fig.6 shows the secondary and 3D structure of Conjugate 5. [0075] Fig.7 shows the secondary and 3D structure of Conjugate 6. [0076] Fig.8 is a scheme for the synthesis of Conjugate 11. [0077] Fig.9 is a scheme for the synthesis of Conjugate 12. [0078] Fig.10 is a scheme for the synthesis of Conjugate 13. [0079] Fig.11 is a scheme for the synthesis of Conjugate 14. [0080] Fig.12 shows images of FAP HT1080 cells from binding studies of FAP-targeting ligands at different concentrations. [0081] Figs.13A-13G show graphical data from in vitro binding studies. Fig.13A is data from a FAP-targeted ligand (Conjugate 12) with HEK-FAP cells. Figs.13B and 13C show data from an enzyme inhibition analysis of the FAP8 base ligand (Fig. 13B) and FAP8-PEG 3 -IP-DOTA (Conjugate 24') (Fig.13C). For FAP and PREP inhibition, FAPi-46 4 was used as a positive control (Fig. 13D). Fig. 13E shows graphical data from a binding analysis of FAP8-PEG 3 -FITC (Conjugate 12) on HEK-hFAP, and Fig.13F shows binding of [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA conjugate (Lu-Conjugate 24') on HEK-hFAP cells. Fig.13G shows graphical data from a binding analysis of FAP8-PEG3-S0456 (Conjugate 58a in Fig.30) on HEK-FAP cells. [0082] Fig. 14 shows binding of [ 64 Cu]-labelled FAP-targeted NOTA conjugate (Cu-Conjugate 18') on HEK-hFAP cells. All assays were performed in triplicate, with SEM bars shown. [0083] Fig. 15A are radio-high performance liquid chromatography (HPLC) chromatograms of FAP8-IP-DOTA (Conjugate 16) and FAP8-DOTA (Conjugate 13) upon chelation with 111 InCl3. [0084] Fig. 15B is a graph of stability data related to the analysis of natural lutetium chelated FAP8-PEG 3 -IP-DOTA ( nat Lu-Conjugate 24') in various formulations as compared to positive controls. [0085] Fig.16 shows radio-images of in vivo studies, with T = tumor, Li = liver, Ki = kidney, and Bla = bladder. [0086] Figs. 17 and 18 are images of HT29 tumor-bearing mice administered FAP8-IP-DOTA conjugate (chelated Conjugate 16), taken at various time points between 4 hours and 168 hours post-conjugate injection. White circles in Fig.18 label the bladder and T = tumor, Li = liver. [0087] Fig. 19 are 111 In-Single Photon Emission Computed Tomography (SPECT) images of HT29 and U87Mg tumor-bearing mice administered FAP8-IP-DOTA conjugate (chelated Conjugate 13) or unlabeled FAP8-DOTA conjugate (Conjugate 13) (far right images), taken at 24 hours post-conjugate injection. White circles in Fig. 19 label the bladder and T = tumor, Ki = kidney. [0088] Fig. 20 are 111 In-SPECT images of HT29 and U87Mg tumor-bearing mice administered FAP8-IP-DOTA conjugate (chelated Conjugate 13) taken from 4 hours to 120 hours post- injection. White circles in Fig.20 label the bladder. [0089] Fig. 21 is a scheme for the synthesis of FAP8 (Compound 10') and FAP8-PEG 3 ligand (Ligand 15'). [0090] Fig. 22 is a scheme for the synthesis of FAP8-DOTA (Conjugate 17'), FAP8-NOTA (Conjugate 18'), and FAP8-NCS-DOTA (Conjugate 19'). [0091] Fig.23 is a scheme for synthesis of FAP8-PEG3-IP-DOTA conjugates (Conjugate 24'), [0092] Fig.24 is a scheme for synthesis of FAP8-IP-NCS-DOTA (Conjugate 25'), FAP8-IP-NCS- NOTA (Conjugate 26'), FAP8-IP-CHX-A-DTPA (Conjugate 27'), and FAP8-IP-NOTA (Conjugate 28'). [0093] Fig. 25 is a scheme for synthesis of FAP8-Tol-DOTA (Conjugate 32') and FAP8-Toly- NOTA (Conjugate 33'). [0094] Fig.26 is a scheme for synthesis of FAP8-akyl-IP-DOTA (Conjugate 38'). [0095] Fig.27 is a scheme for synthesis of FAP8-Pz-IP-DOTA (Conjugate 44') and FAP8-Pz-IP- NCA-DOTA (Conjugate 45'). [0096] Fig.28 is a scheme for synthesis of FAP8-Laurly-DOTA (Conjugate 50'). [0097] Fig.29 is a scheme for synthesis of Compounds 55a-f'. [0098] Fig 30 is a scheme for synthesis of FAP8-S0456 conjugates with different PEG links (Conjugates 59' and 60'). [0099] Fig.31 is a scheme for synthesis of FAP8-PEG3-IRDye800CW (Conjugate 59'). [0100] Fig.32 is a scheme for synthesis of FAP8-OCG dye conjugate (Conjugate 60'). [0101] Fig.33 is a scheme for synthesis of FAP8-FITC conjugate (Conjugate 61'). [0102] Fig.34A is a scheme for synthesis of FAP8-ICG dye conjugate (Conjugate 62'). [0103] Fig.34B is a scheme for synthesis of FAP8-ICG dye conjugates (Conjugates 62', 62a, 62b, 62c, and 62d). [0104] Figs.35A-35C show data from SPECT imaging of [ 111 In] In-FAP8-PEG3-IP-DOTA (In- Conjugate 24') in HEK-FAP tumor bearing mice (Fig. 35A), the biodistribution of [ 177 Lu] Lu- FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') in HEK-FAP tumor bearing mice (n = 4) (Fig.35B), and an analysis of tumor to healthy tissues ratios (Fig.35C). [0105] Figs. 36A and 36B are comparisons of the total absorbed dose coefficient in selected organs (Blood, Heart, lungs, Liver, Spleen, Kidney’s bone marrow and Tumor) in 4T1 and HEK- hFAP tumor bearing mice injected with 5 nmol/mouse FAP8-PEG 3 -IP-DOTA chelated with 7.4 MBq/mouse of 177 Lu (Lu-Conjugate 24') (Fig.36A), and the corresponding Tumor: organ ratios (Fig.36B). [0106] Fig. 37 show data related to the efficacy of [ 177 Lu] Lu-FAP8-IP-DOTA treatment (Lu- Conjugate 24') of MDA-MB-231, KB, and HT29 cancers in tumor-bearing mice (control – A; treatment group – B; n = 5). Treatment groups were treated with 37.0 MBq of [ 177 Lu] Lu-FAP8- IP-DOTA; control groups were treated with PBS. [0107] Fig. 38 show data related to the efficacy of [ 177 Lu] Lu-FAP8-IP-DOTA treatment (Lu- Conjugate 24') of 4T1 cancer in tumor-bearing mice (n = 5) treated with three different doses of [ 177 Lu] Lu-FAP8-IP-DOTA: 37.0 MBq, or 18.5 MBq, or 9.25 MBq). Controls groups were treated with PBS. [0108] Figs. 39A and 39B show SPECT/CT imaging of 111 In-FAP8-PEG 3 -IP-DOTA (In- Conjugate 24') in HT29 tumor bearing mice as a function of time. The dark arrows indicate tumor, the light arrows indicate liver, and fluorescence is highlighted using white circles. [0109] Figs. 40A and 40B show SPECT/CT imaging of 111 In-FAP8-PEG3-IP-DOTA (In- Conjugate 24') in MDA-MB-231 tumor bearing mice as a function of time. The dark arrows indicate tumor, the light arrows indicate liver, and fluorescence is highlighted using white circles. [0110] Figs. 41A and 41B show SPECT/CT imaging of 111 In-FAP8-PEG 3 -IP-DOTA (In- Conjugate 24') in KB tumor bearing mice as a function of time. The dark arrows indicate tumor, the light arrows indicate liver, and fluorescence is highlighted using white circles. [0111] Fig.42 is mass spectrometry data related to chelation of FAP8-PEG3-NOTA (Conjugate 18') with 64 CuCl2 and the stability thereof. [0112] Fig. 43 is PET images of 64 Cu-FAP8-PEG3-NOTA (Conjugate 18') in U87Mg tumor bearing mice at different time points. The white arrow indicates the tumor. [0113] Fig. 44 shows graphs of dose escalation data of 111 In-FAP8-PEG3-IP-DOTA (In- Conjugate 24') in 4T1 tumor-bearing mice measured at 4 hours, 24 hours, and 120 hours post treatment. [0114] Figs. 45A and 45B show dose escalation data of 177 Lu-FAP8-PEG3-IP-DOTA (Lu- Conjugate 24') in 4Ti tumor bearing mice (n = 3) using different doses of conjugate: 0.3 nmol/mouse (Fig.45A) or 1.0 nmol/mouse (Fig.45B). [0115] Figs. 46A-46D show graphical comparisons of the total absorbed dose coefficient in selected organs from the studies of Figs. 44 and 45. Fig. 46A shows the values from selected organs and tumor in 4TI tumor-bearing mice injected with 0.3 nmol/mouse Lu-Conjugate 24'. Fig. 46B shows the values from selected organs and tumor in 4TI tumor-bearing mice injected with 1.0 nmol/mouse Lu-Conjugate 24'. Figs. 46C and 46D show values from selected organs and tumor in 4TI tumor-bearing mice injected with doses of Lu-Conjugate 24' and their corresponding tumor: healthy tissues ration, respectively. [0116] Fig.47 are representative photomicrographs of 4 µm sections of mouse heart, liver, and kidney tissue stained with H&E. The samples were taken following radiotherapy treatments of a single dose of Lu-Conjugate 24' and treatment with 37 MBq in athymic nu/nu mice bearing HT29 tumor. [0117] Fig.48 are representative photomicrographs of 4 µm sections of mouse heart, liver, and kidney tissue stained with H&E. The samples were taken following radiotherapy treatments of a single dose of Lu-Conjugate 24' and treatment with 37 MBq in athymic nu/nu mice bearing MDA- MB-231 tumor. [0118] Fig.49 are representative photomicrographs of 4 µm sections of mouse heart, liver, and kidney tissue stained with H&E. The samples were taken following radiotherapy treatments of a single dose of Lu-Conjugate 24' and treatment with 37 MBq in athymic nu/nu mice bearing KB tumor. [0119] Fig.50 shows whole body and ex vivo fluorescence imaging of FAP8-PEG3-IRDye800CW (Conjugate 58a in Fig.30) in 4T1 tumor bearing mice at different time points. Circles highlight portions where fluorescence was the strongest, with the M1 and M2 tumors at all time points indicating red on the fluorescence scale, M1 and m2 lungs and liver showing yellow/green on the fluorescence scale at 2 hours, green/blue at 6 hours, and negligible at 12 hours and 24 hours (with the exception that M2 lungs rated green at 12 hours), and both M1 and M2 kidneys rating on the fluorescence scale at both 2 hours and 6 hours, one set of M2 kidneys remaining red at 12 hours, but the remaining kidneys from both M1 and M2 negligible fluorescence at 12 hours and 24 hours post treatment. [0120] Fig. 51 shows images of mice and extracted organs from a competition experiment of FAP8-PEG3-IRDye800CW (Conjugate 58a in Fig.30) in 4T1 tumor bearing mice at 6 hours post injection. Circles indicate portions of highest fluorescence, with the M1 tumor and kidneys indicating red on the fluorescence scale, M3 (comp) kidneys showing partially red on the fluorescence scale, M2 and M3 tumors showing yellow/green on the fluorescence scale, and M2 liver and lungs showing blue on the fluorescence scale. [0121] Fig.52 shows images taken of mice and organs from a dose escalation study for FAP8- PEG 3 -IRDye800CW (Conjugate 61') in KB tumor bearing mice. All tumors showed red on the fluorescence scale for each dose amount, with red fluorescence in the liver at 2.5 nmol/mouse and 1.25 nmol/mouse dosing, but not for the 10 nmol/mouse and 5 nmol/mouse dosing. The kidneys for the 1.25 nmol/mouse dose cohort indicated red on the fluorescence scale, while the others did not. [0122] Fig.53 shows whole body and ex vivo fluorescence imaging of KB tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG3-IRDye800CW (Conjugate 58a in Fig. 30). The tumors of the M1 and M2 targeted mice indicated red on the fluorescence scale, while the M1 and M2 of the competition mice did not (no fluorescence). M1 competition mouse did show red fluorescence in the liver and kidneys, while M2 competition mouse only showed green fluorescence on the liver and blue fluorescence on the kidneys. Both M1 and M2 targeted mice had fluorescence in both liver and kidneys (M1 liver = green; M2 liver = red; M1 kidneys = blue; M2 kidneys = red/yellow). [0123] Fig.54 shows whole body and ex vivo fluorescence imaging of HT29 tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG3-IRDye800CW (Conjugate 58a in Fig.30). The tumors of the M1 and M2 targeted mice indicated red on the fluorescence scale, while the M1 and M2 of the competition mice did not (no fluorescence). Both competition mice exhibited red fluorescence in the liver and slightly less in the kidneys, while both targeted mice exhibited red fluorescence in the liver and kidneys. [0124] Fig. 55 shows whole body and ex vivo fluorescence imaging of U87Mg tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG 3 -IRDye800CW (Conjugate 58a). The tumors of the M1 and M2 targeted mice indicated red on the fluorescence scale, while the M1 and M2 of the competition mice did not (no fluorescence). Both competition mice exhibited red fluorescence in the liver and less in the kidneys (green), while both targeted mice exhibited only green and blue fluorescence, respectively, in the liver and blue in the kidneys. [0125] Fig.56 shows whole body and ex vivo fluorescence imaging of 4T1 tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG3-IRDye800CW (Conjugate 58a). The tumors of the M1 and M2 mice indicated red on the fluorescence scale at both 4-hour and 12-hour time points. Livers of both mice were red at 4 hours, but exhibited no fluorescence at 12 hours. Kidneys of all mice shown exhibited red fluorescence at both time points. [0126] Fig.57 shows whole body and ex vivo fluorescence imaging of 4T1 tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG 6 -IRDye800CW (Conjugate 58b in Fig.30). The tumors of all mice indicated red on the fluorescence scale in the whole body image, but not on the biodistribution image. The livers and kidneys of all mice indicated red on the fluorescence scale in the ex vivo biodistribution image. [0127] Fig.58 shows whole body and ex vivo fluorescence imaging of 4T1 tumor-bearing mice at 4 hours post injection with 5 nmol FAP8-PEG8-IRDye800CW (Conjugate 58c in Fig.30). The tumors of all mice indicated yellow/green on the fluorescence scale in the whole body image, except that the mouse on the far left also had red in the middle. The tumors did not indicate significant fluorescence on the biodistribution image. The livers and kidneys of all mice indicated red on the fluorescence scale in the ex vivo biodistribution image. [0128] Fig.59 shows an optional scheme for synthesizing a conjugate hereof to optimize yield. [0129] While the present disclosure is susceptible to various modifications and alternative forms, exemplary embodiments thereof are shown by way of example in the drawings and are herein described in detail. DETAILED DESCRIPTION [0130] While the concepts of the present disclosure are illustrated and described in detail in the description herein, results in the description are to be considered as exemplary and not restrictive in character; it being understood that only the illustrative embodiments are shown and described and that all changes and modifications that come within the spirit of the disclosure are desired to be protected. [0131] Conjugates [0132] Provided are conjugates of formula I: wherein A has the structure: or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from O, N, and S; R1 and R 2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 3 and R 4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 5 and R 6 are independently selected from group consisting of -H, -OH, -F, -Cl, 69860-04 -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 ) 2 CH-, (CH 3 ) 3 C-, -CH 2 Ph, and substituted -CH 2 Ph; R8-R10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R11 is selected from the group consisting of H, D, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, adamantyl, substituted or unsubstituted aryl, substituted or unsubstituted C 7 -C 20 alkyl aryl, wherein the aryl is: wherein: R 12 and R 16 are independently selected from the group consisting of H, D, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R13, R14 and R15 are independently selected from the group consisting of H, D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from H and CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; and B is an imaging or therapeutic agent. In various embodiments, when R 1 , R 2 , R 5 , and R 6 are hydrogen, R 3 and R 4 are not independently H, D or F. [0133] L can be attached to A at any carbon atom of the functionalized 5- to 10-membered N- containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, a 1° amine, a 2° amine, a functionalized alkyl, or a functionalized cycloalkyl. A can have the formula IV or V:

wherein: R 2 is selected from the group consisting of -H, -D, -OH, -F, -Cl, -Br, I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R5 and R6 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 )2CH- , and (CH 3 )3C-; R11 is selected from the group consisting of

; and R 23 -R26 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -CF 3 , -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH-, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl. [0134] B can comprise a radical of a transforming growth factor beta (TGF ^)/Smad inhibitor, a Wnt/ ^-catenin inhibitor, a vascular endothelial growth factor receptor 1 (VEGFR1) inhibitor, a vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor, a vascular endothelial growth factor receptor 3 (VEGFR3) inhibitor, a fibroblast growth factor receptor 1 (FGFR1) inhibitor, a fibroblast growth factor receptor 2 (FGFR2) inhibitor, a platelet-derived growth factor receptor (PDGFR) inhibitor, a FAK inhibitor, a rho kinases (ROCK) inhibitor, a PDGFR inhibitor, a toll- like receptor (TLR) agonist, an NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) inhibitor, an inhibitor of collagen synthesis, or an angiogenesis inhibitor. [0135] B can comprise a chemotherapeutic agent, an anti-fibrotic agent, an anti-cancer agent, or an immunotherapeutic agent. The anti-cancer agent can be effective against cancer cells, cancer- associated fibroblasts, or factors in the tumor microenvironment. [0136] B can comprise a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope, wherein said metal or isotope is suitable for radio-imaging, PET imaging, SPECT imaging, radiotherapy, or magnetic resonance imaging, a fluorescent imaging agent, a photodynamic imaging agent, an optical imaging agent, a photosensitizer, or a radiosensitizer. The fluorescent imaging agent can be selected from the group consisting of carbocyanine, indocarbocyanine, oxacarbocyanine, thiacarbocyanine and merocyanine, polymethine, coumarine, rhodamine, xanthene, fluorescein, borondipyrromethane (BODIPY), Indocyanine green (ICG), CyS, CyS.S, Cy7, VivoTag-680, VivoTag-S680, VivoTag-S7S0, AlexaFluor660, AlexaFluor680, AlexaFluor700, AlexaFluor7S0, 10 AlexaFluor790, Dy677, Dy676, Dy682, Dy7S2, Dy780, DyLightS47, Dylight647, HiLyte Fluor 647, HiLyte Fluor 680, HiLyte Fluor 7S0, IRDye 800CW, IRDye 800RS, IRDye 700DX, ADS780WS, ADS830WS, ADS832WS and S0456. The fluorescent imaging agent can have a structure selected from:

[0137] The fluorescent imaging agent can have a structure selected from:

[0138] The photosensitizer can have a structure selected from:

[0139] The metal chelating group can be selected from the group consisting of DOTA (1,4,7,10- tetraazacyclododecane-1,4,7,10-tetraacetic acid) or a derivative thereof; TETA (1,4,8,11- tetraazacyclotetradecane-1,4,8,11-tetraacetic acid) or a derivative thereof; SarAr (1-N-(4- Aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]-eicosane- 1,8-diamine or a derivative thereof; NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) or a derivative thereof; NETA (4- [2-(bis-carboxymethylamino)-ethyl]-7-carboxymethyl-[1,4,7]tr iazonan-1-yl) acetic acid or a derivative thereof TRAP (1,4,7-triazacyclononane-1,4,7-tris[methyl(2-carboxyethyl)ph osphinic acid) or a derivative thereof; HBED (N,N0-bis(2-hydroxybenzyl)-ethylenediamine-N,N0-diacetic acid) or a derivative thereof; 2,3-HOPO (3-hydroxypyridin-2-one) or a derivative thereof; PCTA (3,6,9,15-tetraazabicyclo[9.3.1]-pentadeca-1(15),11,13-trien e-3,6,9,-triacetic acid) or a derivative thereof; DFO (desferrioxamine) or a derivative thereof; DTPA (diethylenetriaminepentaacetic acid) or a derivative thereof; OCTAPA (N,N0-bis(6-carboxy-2-pyridylmethyl)-ethylenediamine- N,N0-diacetic acid) or a derivative thereof; H2-MACROPA (N,N'-bis[(6-carboxy-2- pyridipmethyl]-4,13-diaza-18-crown-6) or a derivative thereof; S-2-(4-Isothiocyanatobenzyl)- 1,4,7,10-tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA) or a derivative thereof; H 2 dedpa (1,2-[[carboxy)-pyridin-2-yl]-methylamino]ethane or a derivative thereof; and EC20- head comprising β-l-diaminopropionic acid, aspartic acid, and cysteine. [0140] The metal chelating group can be bound to 11 C, 13 C, 13 N, 15 O, 18 F, 32 P, 44 Sc, 47 Sc, 52 Mn, 5 5 Co, 60 Co, 64 Cu, 6 7Cu, 67 Ga, 68 Ga, 86 Y, 89 Sr, 89 Zr, 90 Y, 99m Tc, 111 In, 114m In, 117m Sn, 123 I, 124 I, 125 I, 1 31 I, 149 Tb, 153 Sm, 152 Tb, 155 Tb, 161 Tb, 169 Er, 177 Lu, 186 Re, 188 Re, 211 At, 212 Pb, 212 Bi, 213 Bi, 223 Ra, 2 24 Ra, 225 Ab, 225 Ac, or 227 Th. B can comprise a structure selected from the group consisting of

[0141] The metal chelating group can have a structure selected from:

[0142] In certain embodiments, B comprises a metal chelating group optionally bound to a metal, or a group covalently bound to an isotope comprising a radiosensitizer. The radiosensitizer can be selected from:

[0143] The radiosensitizer can be selected from a topoisomerase inhibitor (e.g., camptothecin, topotecan), hypoxia-activated anthraquinone AQ4N, an alkylating agent (e.g., temozolomide), a drug affecting a DNA repair pathway (e.g., poly(ADP ribose)polymerase inhibitor, AG14,361), and a PRMT5 inhibitor (e.g. JNJ-64619178). [0144] In certain embodiments, B comprises a chemotherapeutic agent. The chemotherapeutic agent can be selected from:

[0145] B can comprise an anti-cancer agent. The anti-cancer agent can be effective against cancer cells, cancer-associated fibroblasts, or factors in the tumor microenvironment. The anti-cancer agent can be selected from: [0146] L of the conjugate can be a bifunctional or trifunctional linker. L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: [0147] L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or can comprise a moiety of the formula: . [0148] L can be or can comprise a moiety of the formula:

[0149] L can be or can comprise a moiety of the formula: wherein n is an integer from 0 to 20. [0150] L can be or can comprise a moiety of the formula:

wherein n is an integer from 0 to 20. [0151] L can be a linker that can be cleaved. L can be cleaved reductively, oxidatively, or enzymatically. L can comprise an oxime ester. L can comprise a hydrazone. L can comprise polyethylene glycoln (PEGn), wherein n = 0-36. L can comprise PEGn, wherein n = 1-36. L can comprise a peptide, a peptidoglycan, an alkyl, or a sugar. [0152] L can be or can comprise: [0153] L can be or comprise a structure selected from: wher ein: R 27 and R 28 are independently selected from the group consisting of H and C 1 -C 6 alkyl; and Z is an integer from 1 to 8. [0154] L can be or comprise the structure: wherein: R31 is H or C1-C6 alkyl; and R 29a , R 29b , R 30a, and R 30b are independently selected from the group consisting of H and C1-C6 alkyl. [0155] L can be or comprise a structure selected from: wherein: R 27 and R 28 are independently selected from the group consisting of H and C 1 -C 6 alkyl; and Z is an integer from 1 to 8. [0156] Also provided is a pharmacokinetically extended conjugate comprising or having the formula (II): or is a stereoisomer or a pharmaceutically acceptable salt, solvate or hydrate thereof, wherein C is a pharmacokinetic extender. A, B, and L of formula (II) can be or comprise any A, B, or L, respectively, described herein in connection with formula (I). [0157] C can be an albumin binder, a plasma protein binder, or a hapten. Where C is an albumin binder, the albumin binder can be or comprise albumin binding domain 035 (ABD035), albumin binding domain Con (ABDCon), a designed ankyrin repeat protein (DARPin), a disulfide stabilized Fv fragment (dsFv), an anti-albumin antibody CA645, an anti-human serum albumin nanobody, or variable new antigen receptor E06 (VNAR E06). [0158] In certain embodiments, a pharmacokinetically extended conjugate can be or comprise a conjugate having formula (II): A has the structure: wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from the group consisting of O, N, and S; R1 and R 2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 5 and R 6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 )2CH-, (CH 3 )3C-, -CH 2 Ph, and substituted -CH 2 Ph; R 8 -R 10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N3, -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R 11 is selected from the group consisting of H, D, C l -C l0 alkyl, C 3 -C l0 cycloalkyl, adamantyl substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: , wherein: R12 and R16 are independently selected from the group consisting of H, D, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 13 , R 14 and R 15 are independently selected from the group consisting of H, D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from H and CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; B is an imaging or therapeutic agent; and C is a pharmacokinetic extender; or is a stereoisomer or a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0159] C of a conjugate hereof can be or can comprise:

[0160] C can be or can comprise:

, wherein: each of R 12-19 is independently -H, -C 1 -C 6 alkyl, -F, -Cl, -Br, -I, -CN, -CHO, -B(OH)2, -C(O)alkyl, -C(O)aryl-, -C=C-C(O)aryl, -C=C-S(O)2aryl, -CO2H, -SO 3 H, -SO 2 NH 2 , -PO 3 H 2 , or -SO 2 F; and each of R 20 and R 21 is independently -H, -C1-C6 alkyl, -F, -Cl, -Br, -I, -O-C 1-6 alkyl, -CN, -CHO, -B(OH) 2 , -C=C-C(O)aryl, -C=C-S(O) 2 aryl, -CO 2 H, -SO 3 H, -SO 2 NH 2 , -PO 3 H 2 , -SO 2 F, CF 3 , or [0161] C can be or can comprise: [0162] C can be or can comprise: [0163] C can be or can comprise: [0164] C can comprise a radical of PEGn, wherein n = 0-32, a peptide, a petidoglycan, or a saccharide. C can be or can comprise:

[0165] C can be or can comprise: The hapten can be recognized by an autologous antibody. The hapten can be selected from the group consisting of rhamnose, an α-galactosyl moiety, a dinitrophenyl (DNP) moiety, and a trinitrophenyl (TNP) moiety. [0166] L can be or comprise a moiety of the formula: wherein n is an integer from 0 to 10. L can be or comprise a moiety of the formula: , wherein n is an integer from 0 to 10. L can be or comprise a moiety of the formula: [0167] L can be or comprise a moiety of the formula:

wherein n is an integer from 0 to 10. L can be or comprise a moiety of the formula: . L can be or comprise a moiety of the formula: [0168] L can be or comprise a moiety of the formula:

wherein n is an integer from 0 to 20. L can be or comprise a moiety of the formula: wherein n is an integer from 0 to 20. L can be or comprise: [0169] L can be or comprise a structure selected from: R27 and R28 are independently selected from the group consisting of H and C1-C6 alkyl; and Z is an integer from 1 to 8. [0170] L can be or comprise a structure selected from: , wherein: R31 is H or C1-C6 alkyl; and R 29a , R 29b , R 30a, and R 30b are independently selected from the group consisting of H and C1-C6 alkyl. [0171] In certain embodiments, the conjugate has or comprises a structure selected from:

[0172] In certain embodiments, the conjugate has or comprise the structure of wherein n = 1-20. [0173] In certain embodiments, the conjugate has or comprises a structure of

wherein n = 1-5. [0174] In certain embodiments, the conjugate has or comprises a structure selected from:

wherein n = 1-5.

[0175] In certain embodiments, the conjugate has or comprises the structure: , wherein n is 0-20. In certain embodiments, n is 2. In certain embodiments, n is 5. In certain embodiments, n is 7. In certain embodiments, n is 11. [0176] In certain embodiments, the conjugate has or comprises a structure selected from:

wherein n = 1-5. [0177] In certain embodiments, the conjugate has or comprises a structure selected from:

[0178] In certain embodiments, the conjugate has or comprises a structure selected from:

wherein m = 1-10 and p = 1-20. [0179] In certain embodiments, the conjugate has or comprises a structure selected from:

, wherein m = 1-10 and p = 1-20. [0180] In certain embodiments, the conjugate has or comprises a structure of: . [0181] In certain embodiments, the conjugate has or comprises a structure of:

[0182] In certain embodiments, the conjugate has or comprises a structure from:

wherein: n = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging.

[0183] In certain embodiments, the conjugate has or comprises a structure selected from: wherein: p = 1-20; and the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0184] In certain embodiments, the conjugate has or comprises a structure selected from: wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0185] In certain embodiments, the conjugate has or comprises a structure selected from: wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging.

[0186] In certain embodiments, the conjugate has or comprises a structure selected from: wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0187] In certain embodiments, the conjugate has or comprises a structure selected from:

optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0188] In certain embodiments, the conjugate has or comprises a structure selected from:

or is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging.

[0189] In certain embodiments, the conjugate has or comprises a structure selected from:

r or is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein: n = 1-5 and p = 1-20; and the compound is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0190] In certain embodiments, the conjugate has or comprises a structure selected from: or is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein the conjugate, e.g., a compound comprising a chelator, is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0191] In certain embodiments, the conjugate has or comprises a structure selected from: or is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein the conjugate, e.g., a compound comprising a chelator, is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0192] In certain embodiments, the conjugate has or comprises a structure selected from: or is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging.

[0193] In certain embodiments, the conjugate has or comprises a structure selected from: is a pharmaceutically acceptable salt, solvate, or hydrate thereof, wherein the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging. [0194] In certain embodiments, the conjugate has or comprises a structure selected from:

is a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0195] In certain embodiments, the conjugate has or comprises a structure selected from: pharmaceutically acceptable salt, solvate, or hydrate thereof. [0196] In certain embodiments, the conjugate has or comprises a structure selected from:

or is a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0197] In certain embodiments, the conjugate has or comprises a structure selected from: pharmaceutically acceptable salt, solvate, or hydrate thereof. [0198] In certain embodiments, the conjugate has or comprises a structure selected from:

is a pharmaceutically acceptable salt, solvate, or hydrate thereof. [0199] In certain embodiments, the conjugate has or comprises a structure selected from:

, wherein n = 0-20. [0200] The conjugates can contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R)- or (S)-. Unless stated otherwise, it is intended that all stereoisomeric forms of the conjugates are contemplated. When the conjugates described herein contain alkene double bonds, and unless specified otherwise, it is intended that this disclosure includes both E and Z geometric isomers (e.g., cis or trans). Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included. The term “geometric isomer” refers to E or Z geometric isomers (e.g., cis or trans) of an alkene double bond. The term “positional isomer” refers to structural isomers around a central ring, such as ortho-, meta-, and para- isomers around a benzene ring. Further, it is understood that replacement of one or more hydrogen atoms with deuterium can significantly lower the rate of metabolism of a drug and, therefore, increase its half-life. [0201] Salts [0202] The compounds and conjugates can be presented as a pharmaceutically acceptable salt. The term “pharmaceutically acceptable salt” refers to those salts whose counter ions can be used in pharmaceuticals. In various embodiments, such salts include, but are not limited to 1) acid addition salts, which can be obtained by reaction of the free base of the parent compound with inorganic acids such as hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, sulfuric acid, and perchloric acid and the like, or with organic acids such as acetic acid, oxalic acid, (D) or (L) malic acid, maleic acid, methane sulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, succinic acid or malonic acid and the like; or 2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, trimethamine, N-methylglucamine, and the like. Pharmaceutically acceptable salts are well-known to those skilled in the art, and any such pharmaceutically acceptable salt is contemplated in connection with the embodiments described herein. [0203] Pharmaceutically acceptable salts can be synthesized from the parent conjugate/compound which contains a basic or acidic moiety by conventional chemical methods. In some instances, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, the disclosure of which is hereby incorporated by reference. [0204] In certain embodiments, it can be desired to modify the conjugate and/or composition synthesis process to optimize yield at production (e.g., when the conjugate is optionally bound to a metal suitable for radio-imaging, radiotherapy, or magnetic resonance imaging). For example, a multiple step process can be utilized to facilitate stability of the conjugate at the pH required for radiolabeling. Accordingly, in certain embodiments, the scheme set forth in Fig. 59 can be emloyed to facilitate a desired yield (e.g., where a conjugate is pH sensitive). While specific conjugates hereof are shown in Fig.59, it will be appreciated that it is not so limited and can be used to prepare any of the conjugates hereof as may be desired. [0205] In various embodiments, suitable acid addition salts are formed from acids which form non-toxic salts. Illustrative examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate salts. [0206] In various embodiments, suitable base salts are formed from bases which form non-toxic salts. Illustrative examples include the arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases also can be formed, for example, hemisulphate and hemicalcium salts. [0207] In each embodiment hereof, it will be understood that the formulae include and represent not only all pharmaceutically acceptable salts of the compounds and conjugates, but also include any and all hydrates and/or solvates of the compound formulae or salts thereof where appropriate. The term “solvate” means a compound, or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate. [0208] Certain functional groups, such as the hydroxy, amino, and like, can form complexes and/or coordination conjugates with water and/or various solvents. Accordingly, the formulae are to be understood to include and represent those various hydrates and/or solvates. Non-hydrates and/or non-solvates of the compounds and conjugates are also included. [0209] Compositions and Routes of Administration [0210] Further provided is a composition comprising a conjugate hereof or a plurality of conjugates hereof (e.g., a conjugate of Formula I or Formula II) and a pharmaceutically acceptable carrier or excipient, such as a composition comprising a conjugate dispersed in a pharmaceutically acceptable liquid carrier. The term "composition" generally refers to any product comprising more than one ingredient, including the conjugate. The compositions can be prepared from isolated conjugates or from salts, solutions, hydrates, solvates, and other forms of the conjugates. [0211] The term means one or more compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The carrier can be an excipient. The choice of carrier can depend on factors such as the particular mode of administration, the effect of the carrier on solubility and stability, and the nature of the dosage form. For example, the carrier can be suitable for parenteral administration. Pharmaceutical compositions suitable for the delivery of compounds as described herein and methods for their preparation may be found, for example, in Remington: The Science & Practice of Pharmacy, 21st edition (Lippincott Williams & Wilkins, 2005). [0212] Pharmaceutically acceptable carriers can include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. Examples of such carriers (or excipients) include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Liquids within which the conjugate can be dispersed include a carrier liquid or an in vivo liquid. By the conjugate being "dispersed" throughout or in a liquid is meant that the conjugate presents as a dispersed phase within the liquid which itself, relative to the conjugate, presents as a continuous liquid medium or phase. The term "liquid" in the context of a liquid carrier is intended to mean a vehicle in which the conjugate is dispersed and which is in a liquid state at least at the temperature of intended use. [0213] A liquid carrier can be made up of one or more different liquids. Suitable pharmacologically acceptable liquid carriers are described in Martin, Remington's Pharmaceutical Sciences, 18 th Ed., Mack Publishing Co., Easton, PA, (1990), and include, but are not limited to, liquids that are sterilized, such as water and oils, including those of petroleum, animal, vegetable, mineral or synthetic origin, such as peanut oil, soya bean oil, mineral oil, sesame oil, and the like. Other liquid carriers include methylene glycol, propylene glycol, polyethylene glycol, polypropylene glycol, ethanol, isopropyl alcohol, and benzyl alcohol. Water or soluble saline solutions and aqueous dextrose and glycerol solutions can be employed as liquid carriers, particularly for injectable solutions. [0214] In practice, the conjugate can be taken up by a subject in vivo, for example, when the conjugate is administered orally or parenterally. In that case, a liquid carrier originally carrying the conjugate can become so dilute in vivo that the surrounding liquid environment throughout which the conjugate is dispersed becomes more representative of an in vivo liquid (i.e., a biological liquid/fluid within the subject) than the original liquid carrier. For example, once administered parenterally, the conjugate might more aptly be described as being dispersed throughout blood rather than an original liquid carrier. Under those circumstances, it can be convenient to refer to the conjugate as being dispersed throughout an in vivo liquid carrier (i.e., a biological liquid/fluid within the subject). [0215] The components of the compositions also can be commingled with the conjugate, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency. [0216] The composition can comprise cremophor, polysorbate, nanoparticles, a polymer, or a hydrogel, for example. In certain embodiments, the pharmaceutical composition comprises a plurality of conjugates and a pharmaceutically acceptable carrier. A pharmaceutically acceptable carrier can include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, and combinations thereof, that are physiologically compatible. One or more other active agents also can be incorporated into a pharmaceutical composition. [0217] In certain embodiments, a pharmaceutical composition further comprises at least one additional pharmaceutically active agent. The at least one additional pharmaceutically active agent can be an agent useful in the treatment of a cancer. In certain embodiments, the at least one additional pharmaceutically active agent can be an agent useful for radiotherapy. In certain embodiments, the at least one additional pharmaceutically active agent can be an agent useful for imaging (e.g., diagnostic imaging). [0218] Pharmaceutical compositions can be prepared by combining one or more conjugates with a pharmaceutically acceptable carrier and, optionally, one or more additional ingredients (e.g., pharmaceutically active ingredients). The formulations can be administered in pharmaceutically acceptable solutions, which can routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients. [0219] Compositions can comprise one or more pharmacologically acceptable additives known to those in the art. For example, the liquid carrier may comprise one or more additives such as wetting agents, de-foaming agents, surfactants, buffers, electrolytes, preservatives, colourings, flavourings, and sweeteners. [0220] The particular nature of a liquid carrier and any additive (if present) can, in part, depend upon the intended application of the composition. A suitable liquid carrier and additive (if present) can be selected for the intended application of the composition. [0221] The composition is suitable for administration to a subject for diagnostic, mapping, and/or therapeutic applications. By "suitable" for administration is meant that administration of the conjugate/composition to a subject will not result in unacceptable toxicity, including allergenic responses and disease states. [0222] For use in therapy or treatment, an effective amount of the conjugate or composition can be administered to a subject by any mode that delivers the conjugate(s) as desired. Administering a composition can be accomplished by any means known to the skilled artisan. Routes of administration include, but are not limited to, intravenous, intramuscular, intraperitoneal, intravesical (urinary bladder), oral, subcutaneous, direct injection, mucosal (e.g., topical to eye), inhalation, and topical. [0223] Colorants and/or flavoring agents can be included. For example, the conjugate can be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents. [0224] Illustrative formats for oral administration include, but are not limited to, tablets, capsules, elixirs, syrups, and the like. [0225] In certain embodiments, a conjugate and/or composition can be administered directly into the blood stream, into muscle, or into an internal organ. Suitable routes for such parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, epidural, intracerebroventricular, intraurethral, intrasternal, intracranial, intratumoral, intramuscular, intranasal, and subcutaneous. Suitable means for parenteral administration include needle (including microneedle) injectors, needle-free injectors, and infusion techniques. Where it is desirable to deliver the compound(s) and/or compositions systemically, the compound(s) and/or composition can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0226] Parenteral formulations are typically aqueous or non-aqueous isotonic sterile solutions that can contain carriers or excipients, such as salts, carbohydrates, anti-oxidants, bactericide, solute and/or buffering agents (preferably at a pH of 3–9) which renders the composition isotonic with the blood of the intended subject, but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle, such as sterile, pyrogen-free water. Such compositions can be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials. [0227] A liquid formulation can be adapted for parenteral administration of a conjugate or composition as described herein. The preparation of parenteral formulations under sterile conditions, for example, by lyophilization under sterile conditions, can readily be accomplished using standard pharmaceutical techniques well-known to those skilled in the art. The solubility of a conjugate can be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents. [0228] Formulations for parenteral administration can be formulated for immediate and/or modified release. A conjugate can be administered in a time-release formulation, for example in a composition which includes a slow-release polymer. The conjugate can be prepared with a carrier that will protect it against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid and polylactic, polyglycolic copolymers (PGLA). Methods for the preparation of such formulations are generally known to those skilled in the art. [0229] Sterile injectable solutions can be prepared by incorporating the conjugate(s), alone or in further combination with one or more other active agents, in the required amount in an appropriate solvent with one or a combination of ingredients described above, as required, followed by filtered sterilization. Typically, dispersions are prepared by incorporating the conjugate(s) into a sterile vehicle, which contains a dispersion medium and any additional ingredients of those described above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying, which yield a powder of the active ingredients plus any additional desired ingredient from a previously sterile-filtered solution thereof, or the ingredients can be sterile-filtered together. [0230] The pharmaceutical composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. [0231] A conjugate, or a pharmaceutical composition comprising a conjugate, can be continuously administered, where appropriate. [0232] Uses and Methods of Use [0233] In view of the above, a method of imaging cells displaying fibroblast activation protein (FAP) on their surfaces (FAP+ cells) in a subject (e.g., a subject in need of imaging) is provided. As used herein, "subject" means either an animal or human subject. "Animal" means primates, livestock animals (including, without limitation, cows, horses, sheep, pigs and goats), companion animals (including dogs, cats, rabbits and guinea pigs), and captive wild animals (including those commonly found in a zoo environment). Laboratory animals such as rabbits, mice, rats, guinea pigs and hamsters are also contemplated as they may provide a convenient test system. Given that FAP homologs have been found in zebrafish and amphibians, i.e., two species of the Xenopus genus, the subject, in certain instances, could be a fish or an amphibian. [0234] The subject can be a human or a mammal of economic importance and/or social importance to humans, for instance, carnivores other than humans (e.g., cats and dogs), swine (e.g., pigs, hogs, and wild boars), ruminants (e.g., cattle, oxen, sheep, giraffes, deer, goats, bison, and camels), horses, and birds including those kinds of birds that are endangered and kept in zoos, and fowl, more particularly domesticated fowl (e.g., poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like) as they are also of economic importance to humans. [0235] The term "subject" does not denote a particular age. Thus, adult, juvenile and newborn subjects are covered. The terms "subject, " "individual" and "patient" may be used interchangeably herein. In certain embodiments, the subject is a mammal. In certain embodiments, the subject is a human. [0236] In certain embodiments, the method of imaging FAP+ cells in a subject comprises (i) administering to the subject one or more conjugates described herein or pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition comprising the same and (ii) obtaining an image of the conjugate(s) (or a portion thereof) bound to FAP on the surfaces of cells displaying FAP, whereupon FAP+ cells in the subject are imaged. [0237] In certain embodiments, a method of imaging FAP+ cells in a subject comprises (i) administering to the subject one or more conjugates described herein or pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition comprising the same and (ii) imaging the conjugate(s) or stereoisomer or pharmaceutically acceptable salt, hydrate, or solvate thereof (or a portion thereof) bound to FAP on the surfaces of cells displaying FAP. The conjugate(s), or pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition employed in the methods hereof can be any of the conjugates, pharmaceutically acceptable salts, hydrates or solvates thereof, or pharmaceutical compositions described herein. In certain embodiments, the conjugate administered to the subject pursuant to the method of imaging is a conjugate of formula I: wherein A has the structure: or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from O, N, and S; R 1 and R 2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 3 and R 4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R5 and R6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 ) 2 CH-, (CH 3 ) 3 C-, -CH 2 Ph, and substituted -CH 2 Ph; R8-R10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N 3 , -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R11 is selected from the group consisting of H, D, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, adamantyl, substituted or unsubstituted aryl, substituted or unsubstituted C 7 -C 20 alkyl aryl, wherein the aryl is: wherein: R 12 and R 16 are independently selected from the group consisting of H, D, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 13 , R 14 and R 15 are independently selected from the group consisting of H, D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from H and CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; and B is an imaging or therapeutic agent. In various embodiments, when R1, R 2 , R5, and R6 are hydrogen, R3 and R4 are not independently H, D or F. It will be understood that the conjugate can comprise any of the conjugate and related embodiments described herein. [0238] In certain embodiments, the imaging is performed after administering the conjugate(s), or pharmaceutically acceptable salt, hydrate or solvate thereof, or pharmaceutical composition. The imaging can be performed by magnetic resonance imaging (MRI), ultrasound, X-ray, optical imaging, Computed Tomography (CT), Single Photon Emission Computed Tomography (SPECT), Positron Emission Tomography (PET), Fluorescence Resonance Energy Transfer (FRET), or any combination of two or more of the foregoing. Accordingly, the conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, and/or composition can be used in conjunction with other in vivo imaging techniques including, but not limited to, ultrasound, X-ray, optical imaging, CT, SPECT, PET and FRET. [0239] The method can further comprise the simultaneous or sequential administration, in either order, of an effective amount of an active agent that is a free radiosensitizer, radioprotector, immunotherapeutic agent, chemotherapeutic agent, anti-cancer drug, or hormone therapeutic agent, or a pharmaceutical composition (e.g., the second pharmaceutical composition) comprising same and a pharmaceutically acceptable carrier or excipient. In certain embodiments, the active agent comprises a radioprotectant. In certain embodiments, the radioprotectant active agent comprises Lys. [0240] The subject can have cancer and the cells, the tissue, or the organ with cancer can be imaged, whereupon cancer is detected. The subject can have a tumor with a microenvironment, and the method can further comprise obtaining a map of the microenvironment of the tumor. The subject can have a tumor, and the method can further comprise obtaining a measurement of a gross target volume and/or a clinical target volume for treatment. [0241] As used herein, the expression "tumor microenvironment" refers to a heterogeneous population of non-cancerous cells surrounding and/or infiltrating a tumor, which are essential to the functionality, physiology and metastasis of the tumor. The skilled person will appreciate that the tumor microenvironment comprises a range of different cell types that may differ based on the size, location, type and stage of a tumor, illustrative examples of which include fibroblasts, pericytes, adipocytes, mesenchymal stromal cells (MSCs), cancer cells and endothelial cells, and combinations thereof (such as pericytes and endothelial cells). While the cells of the tumor microenvironment can be non-cancerous, tumors can recruit and/or regulate such cells to provide a favorable environment to facilitate cancer growth. Accordingly, cells comprised within the tumor microenvironment may be referred to as "cancer-associated" or "tumor-associated." [0242] As the conjugates and compositions hereof can be used to label the cancer cells (by binding, for example, FAP8), the methods hereof can be used to visualize, characterize, monitor and facilitate treatment of a cancer or other disease. [0243] When the bone in the subject is imaged (either in connection with the method or separate therefrom), the method can further comprise diagnosing whether the subject has cancer. [0244] When the subject has been treated for cancer and the tumor site (e.g., tumor microenvironment) in the subject is imaged, the method can further comprise assessing or monitoring the efficacy of treatment. For example, the conjugates and/or compositions can be used to monitor tumor or lesion growth and proliferation quantitatively in vivo. In certain embodiments, a method of monitoring a progression of a cancer in a subject is provided, comprising administering a conjugate, a pharmaceutically acceptable salt, solvate, hydrate, or stereoisomer thereof, or a pharmaceutical composition comprising the conjugate or a pharmaceutically acceptable salt, solvate, hydrate, or stereoisomer thereof to a subject. Such method can further comprise imaging the cancer of the subject. [0245] The subject can be imaged periodically over the course of a therapeutic treatment, and a practitioner can then compare the images and/or otherwise quantify lesion or cancer growth to determine therapeutic efficacy (e.g., if there is a differential killing effect of the cancer cells over the course of the therapeutic treatment, or a relative increase in lesion size or cancer growth). Accordingly, a method is provided for determining a likelihood of success of a therapeutic treatment in a subject. In certain embodiments, the method further comprises assessing or monitoring efficacy of a treatment administered to the subject. [0246] Therapeutic and diagnostic applications suitable for treatment of a condition experienced by the subject can also be employed in combination with the imaging method hereof. Non-limiting examples of suitable therapeutic or diagnostic applications include MRI, MRI guided external beam radiotherapy, MRI guided focal ablation, MRI/Ultrasound fusion focal ablation, MRI guided biopsy, MRI/Ultrasound fusion guided biopsy, MRI guided surgery, MRI guided brachytherapy, and MRI guided infrared camera guided biopsy or therapy. In certain embodiments, the method further comprises administering radiotherapy to the subject, wherein the radiotherapy is administered before, concurrent with, or sequential to administering the conjugate, stereoisomer or a pharmaceutically acceptable salt, hydrate, or solvate of the conjugate, or the composition to the subject. [0247] As noted above, the conjugate/composition comprising one or more conjugates can allow for the detection of cells expressing FAP, such as cells within the tumor microenvironment (e.g., tumor-associated stromal cells and cancer cells) associated with solid tumors. The solid tumors and/or cancer can comprise prostate cancer, glioblastoma, pancreatic cancer, colorectal cancer, breast cancer and lung cancer, for example. By specifically binding to FAP expressed by the cells of the tumor microenvironment, the conjugate can be useful for the identification of the boundaries and margins of tissue affected by cancer (i.e., tumor mapping). It is also contemplated herein that the conjugate(s) and compositions can be useful for the detection (i.e., diagnosis) of cancer or as part of the treatment of cancer. For example, the conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions can be used for tumor mapping. In certain embodiments, tumor mapping is performed prior to the commencement of treatment, such as focal therapy, radiotherapy, proton therapy or brachytherapy. Furthermore, by accurately mapping a tumor, including regions of the tumor microenvironment, surgical resection of the tumor can be performed with more accuracy to limit undesirable side effects and minimising the risk of suboptimal debulking of the tumor mass. [0248] When the conjugates, pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or pharmaceutical compositions comprising the conjugate(s) are administered for imaging, i.e., MRI, the conjugate(s) pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions can be administered by any suitable route including, for example, intravenously, intraperitoneally, subcutaneously, intracranially, intradermally, intramuscularly, intraocularly, intrathecally, intracerebrally, and intranasally. Typically, the conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions are administered intravenously or orally. The conjugates and compositions are administered orally for gastrointestinal scans. The conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions comprising them can be administered intratumorally or peritumorally. [0249] The amount of conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions administered can, in certain embodiments, be the smallest amount sufficient to generate a clinically useful image. Amounts of currently available contrast agents can be used as a guide in determining the amounts of the conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions to be used. [0250] In certain embodiment, the methods hereof further comprise treating the subject, or having the subject treated, for cancer. For example, the method can further comprise administering an effective amount of a treatment for cancer (e.g., a second anti-cancer therapy) at a site where the conjugate accumulates. The treatment can be any suitable treatment, such as surgery, radiotherapy, brachytherapy, photodynamic therapy, photothermal therapy, focal ablation therapy including cryoablation, focal laser ablation and high-frequency ultrasound ablation, chemotherapy, and immunotherapy. [0251] Also in view of the above, several methods of treatment are provided. In an embodiment, a method of treating cancer in a subject is provided. In another embodiment, a method of treating fibrosis in a subject is provided. In yet another embodiment, a method of treating an inflammatory disease or disorder in a subject is provided. The method can comprise administering to the subject an effective amount of an above-described conjugate(s), pharmaceutically acceptable salts, hydrates, or solvates thereof, and/or compositions comprising the same. [0252] In certain embodiments, the conjugate administered to the subject pursuant to the method of treatment is a conjugate of formula I: wherein A has the structure: or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein: represents a functionalized 5- to 10-membered N-containing aromatic or non-aromatic mono- or bi-cyclic heterocycle, which optionally further comprises 1-3 heteroatoms selected from O, N, and S; R1 and R 2 are independently selected from the group consisting of -H, -D - OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R3 and R4 are independently selected from the group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 5 and R 6 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; R 7 is selected from the group consisting of H, D, OH, CH 2 =, -CH 3 , CH 3 CH 2 -, (CH 3 )2CH-, (CH 3 )3C-, -CH 2 Ph, and substituted -CH 2 Ph; R 8 -R 10 are independently selected from group consisting of -H, -OH, -F, -Cl, -Br, -I, -NO 2 , -SO 3 H, -SO 2 NH 2 , -NH 2 , -N3, -NH=NH, -C 1-6 alkyl, -O-C 1-6 alkyl, and -S-C 1-6 alkyl; and R 11 is selected from the group consisting of H, D, C l -C l0 alkyl, C 3 -C l0 cycloalkyl, adamantyl, substituted or unsubstituted aryl, substituted or unsubstituted C7-C20 alkyl aryl, wherein the aryl is: wherein: R12 and R16 are independently selected from the group consisting of H, D, halogen, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 13 , R 14 and R 15 are independently selected from the group consisting of H, D, halogen, -OMe, C 1 -C 3 alkyl, C 1 -C 3 alkoxy, -CF 3 , and -C(=O)-OR 23 , wherein R 23 is selected from the group consisting of H, D, halogen, C 1 -C 4 alkyl, and C 1 -C 3 alkoxy; R 17 , R 18 , R 20 , and R 21 are independently selected from H and CH 3 ; and R 19 and R 22 are independently selected from the group consisting of phenyl, dimethoxyphenyl, and aryl; L is a bifunctional or trifunctional linker; and B is an imaging or therapeutic agent. In various embodiments, when R1, R 2 , R5, and R6 are hydrogen, R3 and R4 are not independently H, D or F. It will be understood that the conjugate can comprise any of the conjugate and related embodiments described herein. [0253] The therapeutic regimen for the treatment of a disease state (e.g., cancer, fibrosis, an inflammatory disease or disorder, etc.) can be determined by a person skilled in the art and will typically depend on factors including, but not limited to, the type, size, stage and receptor status of a tumor (e.g., with cancer) in addition to the age, weight and general health of the subject. Another determinative factor can be the risk of developing recurrent disease. For instance, for a subject identified as being at high risk or higher risk or developing recurrent disease, a more aggressive therapeutic regimen can be prescribed as compared to a subject who is deemed at a low or lower risk of developing recurrent disease. Similarly, for a subject identified as having a more advanced stage of cancer, for example, stage III or IV disease, a more aggressive therapeutic regimen can be prescribed as compared to a subject that has a less advanced stage of cancer. [0254] The terms "treat," "treatment," and "treating" refer to any and all uses which remedy a condition or symptom, or otherwise prevent, hinder, retard, abrogate or reverse the onset or progression of cancer or other undesirable symptoms in any way whatsoever. Thus, the term "treating," and the like, is to be considered in its broadest possible context. For example, treatment does not necessarily imply that a subject is treated until total recovery or cure. In conditions that display or are characterized by multiple signs or symptoms, the treatment need not necessarily remedy, prevent, hinder, retard, abrogate or reverse all signs or symptoms, but can remedy, prevent, hinder, retard, abrogate or reverse one or more signs or symptoms. [0255] The expression "therapeutically effective amount" means the amount of conjugate when administered to a mammal, in particular a human, in need of such treatment, is sufficient to treat cancer. The precise amount of conjugate to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the subject. [0256] "Administration" of the conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, and/or composition to a subject is meant that the conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, or composition is presented such that the conjugate(s) and/or pharmaceutically acceptable salts, hydrates, or solvates thereof can be transferred to the subject. There is no particular limitation on the mode of administration, but this will generally be by way of oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intracerebrally, intranasally, intrathecal, and intraspinal), inhalation (including nebulisation), topical, rectal and vaginal modes. The conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, and/or composition can also be administered directly into a tumor and/or into tissue adjacent one or more segments of a tumor or administered directly into blood vessels. [0257] The conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, and/or composition can be administered in, as appropriate, a treatment or diagnostic effective amount. A treatment or diagnostic effective amount includes an amount which, when administered according to the desired dosing regimen, achieves a desired therapeutic or diagnostic effect, including one or more of: alleviating the symptoms of, preventing or delaying the onset of, inhibiting or slowing the progression of, diagnosing, or halting or reversing altogether the onset or progression of a particular condition being treated and/or assessed. As used herein, “effective amount” means and encompasses both therapeutically effective amount and treatment or diagnostic effective amount. [0258] Suitable dosage amounts and dosing regimens to achieve this can be determined by the attending physician and can depend on the particular condition being treated or diagnosed, the severity of the condition as well the general age, health and weight of the subject. [0259] Depending upon the route of administration, a wide range of permissible dosages are contemplated. The dosing can occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods. Suitable dosages of the particulate material per se can lie within the range of about 0.1 ng per kg of body weight to 1 g per kg of body weight per dosage. The dosage can be in the range of 1 µg to 1 g per kg of body weight per dosage, such as is in the range of 1 mg to 1 g per kg of body weight per dosage. In one embodiment, the dosage can be in the range of 1 mg to 500 mg per kg of body weight per dosage. In another embodiment, the dosage can be in the range of 1 mg to 250 mg per kg of body weight per dosage. In yet another embodiment, the dosage can be in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per body weight per dosage. [0260] Conjugate(s), pharmaceutically acceptable salt, hydrate, or solvate thereof, and/or compositions hereof can be administered in a single dose or a series of doses. For example, dosages may be single or divided and may be administered according to a wide variety of protocols, including q.d. (once a day), b.i.d. (twice a day), t.i.d. (three times a day), or even every other day, once a week, once a month, once a quarter, and the like. In each of these cases it is understood that the effective amounts described herein correspond to the instance of administration, or alternatively to the total daily, weekly, month, or quarterly dose, as determined by the dosing protocol. [0261] In addition to the illustrative dosages and dosing protocols described herein, an effective amount of any one or a mixture of the compounds described herein can be determined by the attending diagnostician or physician by the use of known techniques and/or by observing results obtained under analogous circumstances. In determining the effective amount or dose, a number of factors are considered by the attending diagnostician or physician, including, but not limited to the species of mammal, including human, its size, age, and general health, the specific disease or disorder involved, the degree of or involvement or the severity of the disease or disorder, the response of the individual patient, the particular compound administered, the mode of administration, the bioavailability characteristics of the preparation administered, the dose regimen selected, the use of concomitant medication, and other relevant circumstances. [0262] In certain embodiments, a use of a conjugate, a pharmaceutically acceptable salt, hydrate, or solvate of the conjugate, or a composition hereof in the manufacture of a medicament for the treatment of a disease in a subject is provided. The conjugate can be any compound or conjugate hereof. The disease in the subject can be cancer. The disease in the subject can be fibrosis. The disease in the subject can be an inflammatory disease or disorder. [0263] Any of the conjugates and/or compositions hereof can be for use in the treatment of a subject experiencing and/or having a disease state described herein. The disease state, for example, can be cancer, fibrosis, or an inflammatory disease or disorder. [0264] Those skilled in the art will recognize that numerous modifications can be made to the specific implementations described above. The implementations should not be limited to the particular limitations described. Other implementations may be possible. [0265] While the conjugates and pharmaceutical compositions are illustrated and described in detail in the foregoing description, the same is to be considered as illustrative and not restrictive in character, it being understood that only certain embodiments have been shown and described and that all changes and modifications that come within the spirit of the invention are desired to be protected. [0266] It is intended that that the scope of the present conjugates, compositions, and methods are defined by the following claims. However, this disclosure may be practiced otherwise than is specifically explained and illustrated without departing from its spirit or scope. Those skilled in the art will understand that various alternatives to the embodiments described herein can be employed in practicing the claims without departing from the spirit and scope as defined in the following claims. [0267] Any use of section headings is intended to aid reading of the document and is not to be interpreted as limiting. Further, information that is relevant to a section heading may occur within or outside of that particular section. [0268] All publications, patents, patent application publications, journal articles, textbooks, and other publications referred to in this document are indicative of the level of skill of those in the art to which the disclosure pertains. All such publications are incorporated herein by reference to the same extent as if each individual publication were specifically and individually indicated to be incorporated by reference. In the event of inconsistent usages between this document and those documents so incorporated by reference, the usage in the incorporated reference should be considered supplementary to that of this document; for irreconcilable inconsistencies, the usage in this document controls. [0269] Various techniques and mechanisms will sometimes describe a connection or link between two components. Words such as attached, linked, coupled, connected, and similar terms with their inflectional morphemes are used interchangeably, unless the difference is noted or made otherwise clear from the context. These words and expressions do not necessarily signify direct connections but include connections through mediate components. It should be noted that a connection between two components does not necessarily mean a direct, unimpeded connection, as a variety of other components may reside between the two components of note. Consequently, a connection does not necessarily mean a direct, unimpeded connection unless otherwise noted. [0270] Certain Definitions [0271] As used herein, the following terms and phrases shall have the meanings set forth below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art. [0272] The term "about" or "approximately" means within an acceptable range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g., the limitations of the measurement system. For example, "about" can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. By way of further example, “about” or “approximately” can mean within 90%, within 95%, 99%, 99.5%, 99.9%, 99.99%, or at least about 99.999% or more of a stated value or of a stated limit of a range. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Unless otherwise stated, the term "about" means within an acceptable error range for the particular value, such as ± 1-20%, preferably ± 1-10% and more preferably ±1-5%. [0273] Where a range of values is provided, it is understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both limits, ranges excluding either or both of those limits are also included. [0274] A phrase referring to "at least one of" a list of items refers to any combination of those items, including single members. As an example, "at least one of a, b, or c" is intended to cover: a, b, c, a-b, a-c, b-c, and a-b-c. [0275] The terms “a,” “an,” or “the” are used to include one or more than one unless the context clearly dictates otherwise. The term “or” is used to refer to a nonexclusive “or” unless otherwise indicated. In addition, it is to be understood that the phraseology or terminology employed herein, and not otherwise defined, is for the purpose of description only and not of limitation. [0276] The term “or” is used to refer to a nonexclusive “or” unless otherwise indicated. In addition, it is to be understood that the phraseology or terminology employed herein, and not otherwise defined, is for the purpose of description only and not of limitation. [0277] The term “substituted” as used herein refers to a functional group in which one or more hydrogen atoms contained therein are replaced by one or more non-hydrogen atoms. The term “functional group” or “substituent” as used herein refers to a group that can be or is substituted onto a molecule. Examples of substituents or functional groups include, but are not limited to, a halogen (e.g., F, Cl, Br, and I); an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, aralkyloxy groups, oxo(carbonyl) groups, carboxyl groups including carboxylic acids, carboxylates, and carboxylate esters; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfoxide groups, sulfone groups, sulfonyl groups, and sulfonamide groups; a nitrogen atom in groups such as amines, azides, hydroxylamines, cyano, nitro groups, N-oxides, hydrazides, and enamines; and other heteroatoms in various other groups. [0278] The term “optionally substituted,” or “optional substituents,” as used herein, means that the groups in question are either unsubstituted or substituted with one or more of the substituents specified. When the groups in question are substituted with more than one substituent, the substituents may be the same or different. When using the terms “independently,” “independently are,” and “independently selected from” mean that the groups in question may be the same or different. Certain of the herein defined terms may occur more than once in the structure, and upon such occurrence each term shall be defined independently of the other. [0279] "Oxo" refers to the =O radical. [0280] "Alkyl" generally refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, such as having from one to fifteen carbon atoms (e.g., C1- C 15 alkyl). "Alkyl" is intended to include independent recitations of a saturated "alkyl, " unless otherwise stated. An alkyl can comprise one to thirteen carbon atoms (e.g., C1-C13 alkyl). An alkyl can comprise one to eight carbon atoms (e.g., C 1 -C 8 alkyl). An alkyl can comprise one to five carbon atoms (e.g., C1-C5 alkyl). An alkyl can comprise one to four carbon atoms (e.g., C1- C 4 alkyl). An alkyl can comprise one to three carbon atoms (e.g., C 1 -C 3 alkyl). An alkyl can comprise one to two carbon atoms (e.g., C1-C2 alkyl). An alkyl can comprise one carbon atom (e.g., C1 alkyl). An alkyl can comprise five to fifteen carbon atoms (e.g., C5-C15 alkyl). An alkyl can comprise five to eight carbon atoms (e.g., C 5 -C 8 alkyl). An alkyl can comprise two to five carbon atoms (e.g., C2-C5 alkyl). An alkyl can comprise three to five carbon atoms (e.g., C3-C5 alkyl). In various embodiments, the alkyl group is selected from methyl, ethyl, 1-propyl (n- propyl), 1-methylethyl (iso-propyl), 1-butyl (n-butyl), 1-methylpropyl (sec-butyl), 2- methylpropyl (iso-butyl), 1,1-dimethylethyl (tert-butyl), 1-pentyl (n-pentyl). The alkyl is attached to the rest of the molecule by a single bond. [0281] "Alkoxy" refers to a radical bonded through an oxygen atom of the formula –O-alkyl, where alkyl is an alkyl chain as defined above. [0282] "Alkylene" or "alkylene chain" generally refers to a straight or branched divalent alkyl group linking the rest of the molecule to a radical group, such as having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, i-propylene, n-butylene, and the like. [0283] "Aryl" refers to a radical derived from an aromatic monocyclic or multicyclic hydrocarbon ring system by removing a hydrogen atom from a ring carbon atom. The aromatic monocyclic or multicyclic hydrocarbon ring system contains only hydrogen and from five to eighteen carbon atoms, where at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ^–electron system in accordance with the Hückel theory. The ring systems from which aryl groups are derived include, but are not limited to, benzene, fluorene, indane, indene, tetralin and naphthalene. [0284] "Aralkyl" or "aryl-alkyl" refers to a radical of the formula -R c -aryl, where R c is an alkylene chain as defined above, for example, methylene, ethylene, and the like. The alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain. [0285] "Carbocyclyl" or "cycloalkyl" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which includes fused or bridged ring systems, having from three to fifteen carbon atoms. A carbocyclyl can comprise three to ten carbon atoms. A carbocyclyl can comprise five to seven carbon atoms. The carbocyclyl is attached to the rest of the molecule by a single bond. Carbocyclyl or cycloalkyl is saturated (i.e., containing single C-C bonds only) or unsaturated (i.e., containing one or more double bonds or triple bonds). Examples of saturated cycloalkyls include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. An unsaturated carbocyclyl is also referred to as "cycloalkenyl. " Examples of monocyclic cycloalkenyls include, e.g., cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. Polycyclic carbocyclyl radicals include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. [0286] "Carbocyclylalkyl" refers to a radical of the formula –R c -carbocyclyl, where R c is an alkylene chain as defined above. [0287] "Halo" or "halogen" refers to a bromo, chloro, fluoro or iodo substituent. [0288] "Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halogen radicals, as defined above, for example, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like. [0289] The term "heteroalkyl" refers to an alkyl group as defined above in which one or more skeletal carbon atoms of the alkyl are substituted with a heteroatom (with the appropriate number of substituents or valencies – for example, -CH 2 - may be replaced with -NH- or -O-). For example, each substituted carbon atom is independently substituted with a heteroatom, such as wherein the carbon is substituted with a nitrogen, oxygen, selenium, or other suitable heteroatom. In some instances, each substituted carbon atom is independently substituted for an oxygen, nitrogen (e.g. -NH-, -N(alkyl)-, or -N(aryl)- or having another substituent contemplated herein), or sulfur (e.g. - S-, -S(=O)-, or -S(=O) 2 -). A heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. A heteroalkyl is attached to the rest of the molecule at a heteroatom of the heteroalkyl. A heteroalkyl is a C 1 -C 18 heteroalkyl. A heteroalkyl is a C 1 -C 12 heteroalkyl. A heteroalkyl is a C1-C6 heteroalkyl. A heteroalkyl is a C1-C4 heteroalkyl. Heteroalkyl can include alkoxy, alkoxyalkyl, alkylamino, alkylaminoalkyl, aminoalkyl, heterocycloalkyl, heterocycloalkyl, and heterocycloalkylalkyl, as defined herein. [0290] "Heteroalkylene" refers to a divalent heteroalkyl group defined above which links one part of the molecule to another part of the molecule. [0291] "Heterocyclyl" refers to a stable 3- to 18-membered non-aromatic ring radical that can comprise two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which optionally includes aromatic, fused, and/or bridged ring systems. The heteroatoms in the heterocyclyl radical are optionally oxidized. The heterocyclyl radical is partially or fully saturated. "Heterocyclyl" is intended to include independent recitations of heterocyclyl comprising aromatic and non-aromatic ring structures, unless otherwise stated. The heterocyclyl is attached to the rest of the molecule through any atom of the ring(s). Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, 1,3-benzodioxolyl, 1,4-benzodioxanyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl, 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidinyl, 6,7,8,9-tetrahydro-5H-cyclohepta[4,5]thieno[2,3-d]pyrimidiny l, 5,6,7,8-tetrahydropyrido[4,5-c]pyridazinyl, indolinyl, isoindolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. [0292] "N-heterocyclyl" or "N-attached heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. Examples of such N-heterocyclyl radicals include, but are not limited to, 1-morpholinyl, 1-piperidinyl, 1- piperazinyl, 1-pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl. [0293] "Heteroaryl" refers to a radical derived from a 3- to 18-membered aromatic ring radical that can comprise two to seventeen carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. The heteroaryl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, wherein at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ^–electron system in accordance with the Hückel theory. Heteroaryl includes fused or bridged ring systems. The heteroatom(s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring(s). Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, benzofuranyl, benzooxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzothieno[3,2-d]pyrimidinyl, benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, cyclopenta[d]pyrimidinyl, 6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidinyl, 5,6-dihydrobenzo[h]quinazolinyl, 5,6-dihydrobenzo[h]cinnolinyl, 6,7-dihydro-5H- benzo[6,7]cyclohepta[1,2-c]pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, furo[3,2-c]pyridinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyrimidinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridazinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridinyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-5,6,7,8-tetrahydroquinazolinyl, naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 5,6,6a,7,8,9,10,10a-octahydrobenzo[h]quinazolinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyrazolo[3,4-d]pyrimidinyl, pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, thieno[2,3-d]pyrimidinyl, thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pridinyl, and thiophenyl (i.e. thienyl). [0294] The invention illustratively described herein can be suitably practiced in the absence of any element(s) or limitation(s), which is/are not specifically disclosed herein. Thus, for example, each instance herein of any of the terms "comprising," "consisting essentially of," and "consisting of" may be replaced with either of the other two terms. Likewise, the singular forms "a," "an," and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, where a compound/composition is substituted with “an” alkyl or aryl, the compound/composition is optionally substituted with at least one alkyl and/or at least one aryl. [0295] The terms and expressions employed are used as terms of description and not of limitation. Where certain terms are defined and are otherwise described or discussed elsewhere in the "Detailed Description," all such definitions, descriptions, and discussions are intended to be attributed to such terms. There also is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof. Furthermore, while subheadings may be used in the "Detailed Description," such use is solely for ease of reference and is not intended to limit any disclosure made in one section to that section only; rather, any disclosure made under one subheading is intended to constitute a disclosure under each and every other subheading. [0296] It is recognized that various modifications are possible within the scope of the claimed invention. Thus, although the present invention has been specifically disclosed in the context of preferred embodiments and optional features, those skilled in the art may resort to modifications and variations of the concepts disclosed herein. Such modifications and variations are considered within the scope of the invention as claimed herein. EXAMPLES [0297] The following examples serve to illustrate the present disclosure. The examples are not intended to limit the scope of the claimed invention in any way. [0298] Solvents, reagents and starting materials were purchased from commercial vendors and used as received unless otherwise described. All reactions were performed at room temperature unless otherwise stated. Starting materials were purchased from commercial sources or synthesized according to the methods described herein or using literature procedures. Materials & Methods [0299] Cell Culture [0300] Cancer cell lines, 4T1, KB, HT29, and MDA-MB-231 were purchased from ATCC and cultured as reported in the literature. Roy et al., Theranostics 10: 5778 (2020). HEK-293T cells that were transduced to express high levels of FAP (HEK-hFAP) were generated as described in Mukkamala et al., J Materials Chemistry B 10: 2038-2046 (2022). [0301] Statistical Analysis [0302] Data were analyzed using GraphPad Prism 8 unless otherwise stated. All results are presented as mean ± SE. [0303] Animal Husbandry [0304] Mice used in the studies described herein were provided normal rodent chow and water ad libitum and maintained on a standard 12-hour light-dark cycle. All animal procedures were approved by the Purdue Animal Care and Use Committee. Example 1 Design of High-Affinity FAP-Targeted Ligands [0305] Conjugates 2 and 3 were prepared pursuant to the scheme and using the reagents shown in Fig.1. [0306] Conjugates 5 and 6 were prepared pursuant to the scheme and using the reagents shown in Fig.2. Example 2 Molecular docking of FAP Ligand (Conjugates 2, 3, 5 and 6) with FAP (1Z68) [0307] Induced fit docking (IFD): The standard IFD protocol in the Schrodinger software package was used to dock the ligands of interest into the binding pocket of FAP. First, a receptor grid box was generated by specifying the amino acid residues in FAP reported being involved in binding interactions. The IFD protocol utilized the Glide docking protocol to generate up to 20 poses for each ligand which are further refined using the Prime Refinement module. [0308] The residues within 5Å of ligand poses were refined and the side chains of the residues were optimized. Upon refinement of the binding site after initial docking, the ligands were re- docked into structures that are within 30.0 kcal/mol of the best structure and within the top 20 structures overall. [0309] The standard precision (SP) scoring function was used in the Glide redocking step to get the final docking scores. The three-dimensional (3D) structure, secondary structure, and docking scores for each of Conjugates 1, 2, 3, 5, and 6 are shown in Figs.3-7, respectively. Example 3 Synthesis of FAP-Targeted Ligand with Free Amine (Conjugate 11) [0310] Conjugate 11 was prepared pursuant to the scheme and using the reagents shown in Fig. 8. Example 4 Synthesis of FAP8 Fluorescein Isothiocyanate (FITC)-Conjugate (Conjugate 12) [0311] Conjugate 12 was prepared pursuant to the scheme and using the reagents shown in Fig. 9. Example 5 Synthesis of FAP8 S0456 Conjugate (Conjugate 13) [0312] Conjugate 13 was prepared pursuant to the scheme and using the reagents shown in Fig. 10. Example 6 Additional Synthesis Schemes [0313] Conjugate 14 (FAP8-DOTA) was prepared pursuant to the scheme and using the reagents shown in Fig.11. [0314] FAP8 (Compound 10) and FAP8-PEG 3 ligand (Ligand 15') were prepared pursuant to the scheme and using the reagents shown in Fig.21. [0315] FAP8-DOTA (Conjugate 17'), FAP8-NOTA (Conjugate 18'), and FAP8-NCS-DOTA (Conjugate 19') were prepared pursuant to scheme and using the reagents shown in Fig.22. [0316] FAP8-PEG3-IP-DOTA conjugates (Conjugate 24') was prepared pursuant to the scheme and using the reagents shown in Fig.23. [0317] FAP8-IP-NCS-DOTA (Conjugate 25'), FAP8-IP-NCS-NOTA (Conjugate 26'), FAP8-IP- CHX-A-DTPA (Conjugate 27'), and FAP8-IP-NOTA (Conjugate 28') were prepared pursuant to the scheme and using the reagents shown in Fig.24. [0318] FAP8-Tol-DOTA (Conjugate 32') and FAP8-Toly-NOTA (Conjugate 33') were prepared pursuant to the scheme and using the reagents shown in Fig.25. [0319] FAP8-akyl-IP-DOTA (Conjugate 38') was prepared pursuant to the scheme and using the reagents shown in Fig.26. [0320] FAP8-Pz-IP-DOTA (Conjugate 44') and FAP8-Pz-IP-NCA-DOTA (Conjugate 45') were prepared pursuant to the scheme and using the reagents shown in Fig.27. [0321] FAP8-Laurly-DOTA (Conjugate 50') was prepared pursuant to the scheme and using the reagents shown in Fig.28. [0322] The Compounds 55a-f' were prepared pursuant to the scheme and using the reagents shown in Fig.29. [0323] FAP8-S0456 conjugates with different PEG links (Conjugates 59' and 60') were prepared pursuant to the scheme and using the reagents shown in Fig.30. [0324] FAP8-PEG 3 -IRDye800CW (Conjugate 58a) was prepared pursuant to the scheme and using the reagents shown in Fig.31. [0325] FAP8-OCG dye conjugate (Conjugate 60') was prepared pursuant to the scheme and using the reagents shown in Fig.32. [0326] FAP8-FITC conjugate was prepared pursuant to the scheme and using the reagents shown in Fig.33. [0327] FAP8-ICG dye conjugate (Conjugate 62) was prepared pursuant to the scheme and using the reagents shown in Fig.34A. [0328] FAP8-ICG dye conjugates (Conjugates 62', 62a, 62b, 62c, and 62d) were prepared pursuant to the scheme and using the reagents shown in Fig.34B. Example 7 Synthesis of FAP8 (Compound 10') and FAP8-PEG3 Ligand (Ligand 15') [0329] Compound 10' and Ligand 15' were prepared pursuant to the scheme and using the reagents shown in Fig.21 pursuant to the below-described steps. [0330] Synthesis of tert-butyl (S)-4,4-difluoro-2-formylpyrrolidine-1-carboxylate (3): [0331] Step-i: LiBH4 (1.1 eq, 4M in THF) was added, slowly dropwise, to a stirred solution of 1- (tert-butyl) 2-methyl (S)-4,4-difluoropyrrolidine-1,2-dicarboxylate (compound 1') (1g, 3.77 mmol) in tetrahydrofuran (THF) (10 mL) at 0 o C. The reaction mixture was transferred to room temperature and stirred there for 1 hour, then evaporated under vacuum to obtain a residue. The residue was redissolved in CH 2 Cl2 (100 mL) and thereafter extracted by saturated aqueous sodium bicarbonate solution (50 mL). The organic layer was separated and evaporated under vacuum, and a crude residue was obtained and purified by column chromatography using ethyl acetate hexanes as mobile phase, to provide the desired compound (compound 2') 850 mg, 95% as white gummy liquid. [0332] Step-ii: Dess-Martin periodinane (4.92 g, 11.60 mmol) was added, portion wise, to a solution of tert-butyl 4,4-difluoro-2-(hydroxymethyl) pyrrolidine-l-carboxylate (compound 2') (2.5 g, 10.55 mmol) in dichloromethane (DCM) (30 mL) at 0 °C. After complete addition, the reaction was warmed to room temperature and stirred for 2 hours. Saturated NaHCO 3 was added, and the layers separated using a phase separator. The DCM was removed in vacuo to give a clear oil which was purified by combi flash using ethyl acetate and hexanes as mobile phase to provide the desired aldehyde 3' (2.25g, 90%) as white gummy liquid. [0333] Synthesis of 4-(isocyanomethyl)-1,2-dimethoxybenzene (compound 5'): [0334] Et 3 N (5.0 eq, 12.82 mmol, 1.68 mL) was added to a stirred solution of compound 4' (0.5g, 2.56mmol) in dry DCM (1.5 mL) at 0 o C, followed by the addition of POCl3 (1.5 eq, 3.84 mmol, 0.24 mL). The reaction mixture was continued at same temperature for 1hour, with progress of the reaction monitored by thin layer chromatography (TLC). [0335] After completion of the starting materials as indicated on the TLC plates, the reaction mixture was further diluted with DCM, absorbed on a silica-gel cartridge, and purified using ethylacetae+hexanes as mobile phase to provide the desired compound 5' (750mg, 81%) as a light yellow liquid, which slowly converted to light yellow solid upon storage. [0336] Synthesis of compound 8': [0337] To a mixture of N-Boc-L-prolinal (1 eq.200 mg, 0.851 mmol), N-protected glycine (1 eq, 161mg, 0.851mmol) and isocyanide (1 eq, 162 mg, 0.851 mmol.) were dissolved in anhydrous DCM and stirred for 4 hours. After complete conversion (liquid chromatography-mass spectrometry (LC-MS)) of the starting materials, trifluoroacetic acid (2.0 mL) was added and the mixture was stirred for 1 hour. The volatiles were evaporated under reduced pressure. The oily residue was redissolved in anhydrous DCM and cooled down to 0° C with an ice bath. Triethylamine (2.0 mL) was added dropwise and stirring continued until the full conversion (LC- MS) (less than 2 hours). The liquids were evaporated under reduced pressure, and the mixture was redissolved in DCM and washed 3 times with water. Organic phase was washed with brine, dried over sodium sulfate, and the solvent was evaporated under reduced pressure and obtained crude residue was purified by using combiflash with hexanes + ethyl acetate as mobile phase provided desired α-hydroxyamide. [0338] The product was then used in the next step by dissolving in MeOH +AcOH (1:1) 10% Pd- C was added (100 mg for 1 g of starting material), and the mixture was stirred under hydrogen atmosphere for 6 hours. Thereafter, the reaction mixture was filtered thorough a celite pad and filtrate was evaporated under reduced pressure to obtain a crude residue of azeotrope with EtOH, which was then purified by combiflash using MeOH +DCM to provide amine 8' as orange color solid. [0339] Synthesis of compound 12': [0340] Cs 2 CO 3 (2.65 gm, 7.93 mmol) was added to a solution of compound 9' (500 mg, 2.64mmol) in DMF (10. mL), followed by the addition of tertiary-butyl bromo acetate (7.93 mmol). The reaction mixture was stirred at 55 o C for 4 hours, followed by the addition of potassium hydroxide (KOH) (7.93 mmol) +H2O (5.0 mL) to the same reaction mixture and continued stirring for additional 2 hours. Progress of the reaction was monitored by LC-MS, and the reaction mixture carefully neutralized with 1N HCl and purified by using reverse phase combi flash using (20 mM, P H = 7.0 ammonium acetate buffer) and acetonitrile as mobile phase to provide the desired acid 10' as white solid (650 mg 81%). [0341] Synthesis of compound 13': [0342] PyBOP (411 mg, 0.792 mmol) and N,N-Diisopropylethylamine (DIPEA) (0.22 mL, 1.32 mmol) were added to a stirred solution of compound 12' (200 mg, 0.66 mmol) in anhydrous CH 2 Cl 2 (10.0 mL). Stirring continued stirring there for 10 minutes. Amine 8' (0.66 mmol) was then added to the above reaction mixture and stirring continued there for additional 2 hours. The reaction mixture was diluted with water, then extracted into DCM (2x20 mL). The combined organic extracts were dried over anhydrous sodium sulphate, filtered, and the filtrate was evaporated under reduced pressure. The crude residue was purified by combiflash using MeOH+DCM as mobile phase to provide compound 13' as white solid. [0343] Synthesis of Ligand 15': [0344] Trifluoroacetic acid (TFA) (2.0 mL) was added to a solution of compound 13' (200.0 mg, 0.297mmol) in DCM (5 mL) and the mixture was stirred at room temperature for 2 hours. Thereafter, the reaction mixture was evaporated under reduced pressure and dried under vacuum, and the resulting crude residue was redissolved in DCM (10.0 mL), followed by the addition of PyBOP (1.2 eq) + DIPEA (5.0eq). After 10 minutes of stirring, BocNH(PEG) 3 NH 2 (1.2 eq) was added to the above reaction mixture, and stirring continued there for additional 2 hours. Work up and purification followed the same procedure as described above to provide compound 14' as a white solid. [0345] Finally, compound 14' (1.0 eq) was redissolved in DCM followed by Dess-Martin periodinane (DMP) (3.0 eq), and water (10. eq) was added and stirred at room temperature overnight. The reaction mixture was further diluted with water and extracted into DCM (2x30 mL), the combined organic extracts were dried over anhydrous sodium sulphate and filtered, and the resulting filtrate was evaporated under reduced pressure. The obtained crude residue was then purified by combiflash using MeOH +DCM as mobile phase to provide the desired keto ligand 15' as a white solid. Example 8 Synthesis of FAP8 Conjugates (Conjugates 17', 18', and 19') [0346] TFA (0.5 mL) was added to a stirred solution of ligand 15' (100 mg, 0.1126 mmol) in DCM (1.0 mL) and stirring continued there for 30 minutes. The reaction mixture was then evaporated under reduced pressure to provide the amine 14' as brown color gummy solid, and this amine was used for further steps without purification. [0347] DIPEA (0.11mmol) was added to a stirred solution of Compound 16' (20 mg, 0.0253 mmol) in DMF (500 ul ), followed by the addition of N-hydroxysuccinimide (NHS)-DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) (16.0 mg). The reaction mixtures was continuously stirred at room temperature for an additional 6 hours. Thereafter, the. reaction mixture was diluted with water and purified by ultra-high performance liquid chromatography (U- HPLC) using (A = Ammonium acetate (10 mM, pH = 7.5), B = Acetonitrile for 60 minutes using a 5-35 method. The desired fractions obtained were quickly freezed using liquid nitrogen and lyophilized for 48 hours to provide the desired conjugate FAP8-DOTA (Conjugate 17') as a white solid (20 mg, 68%). [0348] For synthesis and purification of FAP8-NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) (Conjugate 18') and FAP8-N-chlorosuccinimide (NCS)-DOTA (Conjugate 19') conjugates, the same procedure was followed as described for the FAP8-DOTA conjugate (Conjugate 17') using respective reagents such as NCS-NOTA and p-NCS-Bn-DOTA, respectively. Example 9 Synthesis of FAP8-PEG3-IP-DOTA Conjugates (Conjugate 24') [0349] To a stirred solution of 4-(4-iodophenyl)butanoic acid (Compound 20') (1g, 3.460 mmol) in DCM (15 mL) were added PyBOP (1.2 eq) then DIPEA (2.0 eq), with continued stirring at room temperature for 10 minutes, then the (S)-5-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)- 5-carboxypentan-1-aminium chloride 21' (1.39 gm, 3.460 mmol) and DIPEA (2.0eq) were added to the above reaction mixture. Stirring was again continued there for an additional 4 hours, with progress of the reaction monitored by LC-MS. The reaction mixture was diluted with water and extracted into DCM (2x30 mL). Combined organic extracts were evaporated under reduced pressure and the obtained crude residue was purified by using combiflash with DCM +MeOH as mobile phase to yield the desired compound coupled product (Conjugate 22') (2.0 gm, 90%) as white solid. [0350] Finally, PyBOP (1.2 eq) and DIPEA (2.0 eq) were added to a stirred solution of acid Conjugate 22' (50 mg, 0.0781 mmol) in DCM (2.0 mL), wherein the reaction mixture was stirred there for 10 minutes. Thereafter, amine 16' (0.0781 mmol) was added to the above reaction mixture and stirring was continued for an additional 2 hours. The reaction mixture was then diluted with water (20 mL), extracted into DCM (2x 20 mL), and the combined organic extracts were evaporated under reduced pressure to obtain a crude residue that was thereafter purified by combiflash using silicagel column and MeOH+DCM as mobile phase to provide Conjugate 23a' as a white solid. [0351] Then Conjugate 23a' (50.0 mg, 0.035mmol) was dissolved in DCM (500 uLmL) + diethylamine (500 ul) or 20% Piperdine in DMF and stirred at room temperature for 1 hour. The reaction mixture was evaporated under reduced pressure and the obtained crude residue was washed with diethyl ether, dried under reduced pressure, and obtained Amine 23b' was re- dissolved in DMF (1.0 mL) followed by DIPEA (5.0 eq). Thereafter, NHS-DOTA (1.0 eq) were added and the reaction mixture was stirred at room temperature for 6 hours. Workup and purification followed the same procedure as described in case of Conjugates 17'-19' synthesis as described in Example 8 to provide Conjugate 24' as a white solid. Example 10 Confocal Binding Studies of FAP-Targeting Ligand [0352] HT1080-FAP cells (1,000,000 cells/well) were seeded in 4 well confocal plates. The cells were allowed to grow as a monolayer over 24 hours at 37 o C and incubated with various concentrations of the Conjugate 12 (Fig.9), ranging from 0.5 nM (lowest) to 25 nM (highest) in 1% fetal bovine serum (FBS) in phosphate buffered saline (PBS) for 1 hour at 37 o C. The unbound fluorescence was removed by washing cells with 1% FBS (3x500 μL), which left the cells in 500 µl of 1% FBS. [0353] Images were acquired of the cells with confocal microscopy pursuant to known protocols (Fig.12). [0354] Additionally, FAPα, PREP and DPP4 recombinant enzymes were purchased from R&D Systems and enzyme buffer was obtained from BPS Bioscience. H-Gly-Pro-AMC and Z-Gly-Pro- AMC were purchased from Bachem Americas, Inc. (Torrance, CA). Increasing concentrations of FAP8 base ligand (Compound 10 (Fig. 21)), FAP8-PEG3-IP-DOTA ligand (Conjugate 24') (0, 0.04, 0.4, 4, 40, 400, 4000, 40000 nM) 25µL + Z-Gly-Pro-AMC for FAP and PREP or H-Gly- Pro-AMC for DPP-IV (5 µL, 1.0 mM) in enzyme buffer (60 µL) were added purified human enzymes FAP (50 ng, catalog number: 3715-SE), PREP (25 ng, catalog number: 4308-SE) and DPP-IV (10 ng, catalog number: 9168-SE-010)) in 10 µL of enzyme buffer then incubated at room temperature for 60 minutes. Fluorescence was then measured using a Cary Eclipse Fluorescence Spectrophotometer at an excitation wavelength of 380 nm and emission wavelength of 450 nm. All assays were performed in triplicate. Example 11 Analysis of FAP Ligand Affinity and Specificity [0355] HEK-FAP cells grown in a monolayer in well plates were incubated with various concentrations of FAP-targeted conjugates, either in the presence or absence of excess of competition ligand (i.e., ligand without dye). [0356] Briefly, HEK-hFAP cells (200,0000)/well were seeded into amine-coated 24 well plates to ensure cell adherence. Upon formation of a monolayer, cells were incubated at 4 o C for 1 hour with a desired concentration of: (a) a FAP-targeted FITC conjugate FAP8-PEG 3 -FITC (Conjugate 12) (Figs.13A and 13E); (b) a FAP base ligand (Fig.13B); (c) unlabelled FAP8-PEG3-IP-DOTA (Conjugate 24') (Fig. 13C); (d) FAPi-46, a positive control from the literature (Fig. 13D); (e) 111 In- or 177 Lu-labelled FAP8-PEG3-IP-DOTA (Conjugate 24') (Fig. 13F); or (f) FAP8-PEG3- S0456 conjugate (Conjugate 58a) (Fig. 13G) in the presence or absence of excess of unlabelled FAP8 ligand (ligand without dye; competition). Cells were then washed 3x with PBS to remove unbound fluorescence or radioactivity and then dissolved in 300 µl of 1% sodium dodecyl sulfate (SDS). [0357] The resulting solution was then transferred into a 96 well black plate (for quantitation of FAP8-PEG3-FITC (Conjugate 12) fluorescence) or gamma counting tubes (for 111 In- or 177 Lu- labelled FAP8-PEG3-IP-DOTA (Conjugate 24') counting), and fluorescence was measured using a fluorescence NeO2 Plate Reader set (ʎex= 488 nm and ʎem= 520 nm) while radioactivity was counted using a HidexAMG gamma counter. Cell-bound fluorescence or radioactivity was plotted against various concentrations and the apparent K D was determined using one-site binding (hyperbola) curve fitting program in GraphPad prism7. All experiments were performed in triplicate. [0358] The above protocol was then repeated for a [ 64 Cu]-labelled FAP-targeted NOTA conjugate (Cu-Conjugate 18') alone as compared to a [ 64 Cu]-labelled FAP-targeted NOTA conjugate (Cu- Conjugate 18') + FAP8-NOTA (in the presence of a competition ligand) on HEK-FAP cells. Briefly, HEK-hFAP cells (200,0000)/well were seeded into amine-coated 24 well plates to ensure cell adherence. Upon formation of a monolayer, cells were incubated at 4 o C for 1 hour with a desired concentration of: (a) a [ 64 Cu]-labelled FAP-targeted NOTA conjugate (Cu-Conjugate 18'); or (b) a [ 64 Cu]-labelled FAP-targeted NOTA conjugate (Cu-Conjugate 18') + FAP8-NOTA (Fig. 14). Example 12 In Vivo Specificity Studies [0359] Mice were inoculated with 3×10 5 HT-29 cells (a human colorectal adenocarcinoma cell line) subcutaneously. When tumors grew to about 300-400 mm 3 , the mice were randomly assigned to 3 groups (n = 3 per group) and treated with an injection Conjugate 24' chelated with 111 InCl 3 ( 5 nmol/mouse) or 177 Lu (5 nmol/mouse). Radio-labeling purity was greater than 95% in each case. Thereafter, the mice were subjected to ~ 450 µCi radioactivity and infused with intravascular lysine injection. [0360] The mice were assessed using 111 In-single-photon emission computed tomography (SPECT) at various time points between 4 hours and 168 hours post-injection (Figs.17-18). [0361] As shown in Fig.16, the conjugates were highly specific to the tumors. There was some liver uptake at 4 hours post-conjugate injection and a significant amount of bladder uptake (the usual excretion pathway; bladders identified in Fig.16 by “Bla” and with a white circle). [0362] In vivo competition studies were also conducted. One group of mice were inoculated with 3×10 5 HT-29 cells subcutaneously, and one group of mice were inoculated with 3×10 5 U87Mg cells (glioblastoma cell line). When tumors grew to about 300-400 mm 3 in each group, the mice were treated with either an injection of Conjugate 13 chelated with 111 InCl3 (5 nmol/mouse), or for the HT-29 mice, 100-fold excess of unlabeled Conjugate 13 (i.e., the competition mouse). Radio-labeling purity was greater than 95% in each case where Conjugate 13 chelated with 111 InCl 3 was administered. Thereafter, the mice were subjected to ~ 450 µCi radioactivity and infused with intravascular lysine injection. [0363] The mice were assessed using 111 In-SPECT at various time points between 4 hours and 120 hours post-injection (Figs.19-20). [0364] As shown in Fig. 19, the conjugates were highly specific to the tumors, even at 4 hours post-injection. There was only bladder uptake (the usual excretion pathway) initially, but it was cleared by 24 hours (see white circles in Fig.19), indicating receptor-mediated uptake. Example 13 Chelation of FAP8-IP-DOTA (Conjugate 24') and FAP8-DOTA (Conjugate 13) with 111 InCl3 [0365] A mixture of FAP8-IP-DOTA (Conjugate 16) or FAP8-DOTA (Conjugate 13) + radioactivity (5 nmol/500 μCi) in ammonium acetate buffer (1.0 M, pH=7) was heated at 90 °C for 10 minutes. Chelation was confirmed by radio-high performance liquid chromatography (HPLC) by dissolving 2 μl of reaction mixture in 10 μl of PBS (Fig.15A). [0366] After chelation was confirmed, 5 mM DTPA (diethylenetriaminepentaacetic acid) (pH = 5) was added to give a final concentration of 200 μM. The reaction mixture was further diluted with 5% ethanol in PBS for in vitro and in vivo experiments. Example 14 Stability Analysis of nat Lu-FAP8-PEG3-IP-DOTA ( nat Lu-Conjugate 24') [0367] As shown in Fig. 15B, a stability analysis of natural lutetium chelated FAP8-PEG 3 -IP- DOTA ( nat Lu-Conjugate 24') was performed in a formulation solution (labelled c), human (labelled a) and mouse plasma (labelled b). Known propantheline was used as positive control (labelled d and e). Intact nat Lu-Conjugate 24' was measured as various time points as indicated in Fig.15B. Example 15 Analysis of RLT Biodistribution [0368] Balb/c mice were inoculated on their shoulders with 1 x 10 5 cells of 4T1 cells, whereas nu/nu mice were inoculated on their shoulders with 5 x 10 6 cells of HT29 (a human colorectal adenocarcinoma cell line), KB (tumor cell line HeLa), MDA-MB-231 (a human breast cancer cell line), or HEK-FAP cells. [0369] For the first study, two different tumor-bearing mice (4T1 and HEK-hFAP) were intravenously injected with [ 111 In] In-FAP8-PEG 3 -IP-DOTA (In-Conjugate 24') (0.74MBq/mouse, 1-10nmol/mouse) or [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') (7.4 MBq/mouse, 0.3 nmol-5 nmol/mouse). [0370] At indicated time points (1 hour, 4 hours, 24 hours, 72 hours, 120 hours, and 168 hours post-injection), mice were assessed and/or euthanized by CO2 asphyxiation and organs of interest were harvested, excess blood was rinsed, dried, weighed, and then analyzed by gamma counter, and the results were decay corrected and presented as the percentage of the injected dose per gram of tissue (%ID/g) ± standard deviation (SD). [0371] Fig.35A shows SPECT imaging of 111 In-FAP8-PEG3-IP-DOTA at the various time points, and Figs. 35B and 35C show graphical data related to the biodistribution of the conjugates and the analysis of tumor to healthy tissue ratios, respectively. Tables 1-8 below provide biodistribution data related to each conjugate and dose studied. In sum, the In-labeled Conjugate 24' targeted the tumors, with minimal off-target concentrations. Table 1. Biodistribution of 111 In-FAP8-PEG 3 -IP-DOTA in 4T1 tumor-bearing mice (1 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 2. Biodistribution of 111 In-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (2.5 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 3. Biodistribution of 111 In-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (5.0 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 4. Biodistribution of 111 In-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (10.0 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 5. Biodistribution of 177 Lu-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (0.3 nmol/mouse)

The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 6. Biodistribution of 177 Lu-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (1.0 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 7. Biodistribution of 177 Lu-FAP8-PEG3-IP-DOTA in 4T1 tumor bearing mice (5.0 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. Table 8. Biodistribution of 177 Lu-FAP8-PEG3-IP-DOTA in HEK-FAP tumor bearing mice (5.0 nmol/mouse) The Values (% ID/g) represent the mean ± SD of data obtained from three animals per cohort. [0372] From the ex vivo biodistribution data obtained from the 111 In/ 177 Lu-FAP8-PEG3-IP-DOTA in 4T1 and HEK-FAP tumor bearing mice, the total absorbed radiation dose coefficients (mGy/MBq) were calculated using OLINDA 2.2.3 software (Tables 9-12). Table 9. 177 Lu-FAP8-PEG3-IP-DOTA dosimetry analysis using OLINDA software (1.0 nmol/mouse and 2.5 nmol/mouse) Table 10. 111 In-FAP8-PEG3-IP-DOTA dosimetry analysis using OLINDA software (5.0 nmol/mouse and 10.0 nmol/mouse)

Table 11. 177 Lu-FAP8-PEG3-IP-DOTA dosimetry analysis using OLINDA software (0.3 nmol/mouse and 1.0 nmol/mouse) Table 12. 177 Lu-FAP8-PEG 3 -IP-DOTA dosimetry analysis using OLINDA software (5 nmol/mouse in 4T1 and 5 nmol/mouse in HEK-FAP) [0373] Figs.36A and 36B show comparison data related to the total absorbed dose coefficient in selected organs (blood, heart, lungs, liver, spleen, kidney’s bone marrow, and tumor). Dose estimates for healthy organs such as blood, heart, lungs, liver, spleen, kidneys, and bone marrow were calculated assuming a 25 g mouse phantom, whereas for tumor dose estimates were calculated with the sphere model. Time-activity curves were fitted to the biodistribution data of each organ individually with exponential functions. [0374] For a second study, HT29 tumor-bearing mice were intravenously injected with either (a) [ 111 In] In-FAP8-PEG 3 -IP-DOTA (In-Conjugate 24') (47.1 MBq/cc) alone (Fig. 39A); or (b) In- Conjugate 24' (47.1 MBq/cc) with unlabeled FAP8 base ligand for competition (Fig. 39B). At indicated time points (1 hour, 4 hours, 24 hours, 72 hours, 120 hours, and 168 hours post- injection), mice were assessed and/or euthanized by CO2 asphyxiation and organs of interest were harvested, excess blood was rinsed, dried, weighed, and then analyzed by gamma counter, and the results were decay corrected and presented as the percentage of the injected dose per gram of tissue (%ID/g) ± standard deviation (SD). In-Conjugate 24' in the HT29 tumor bearing mice had 100-fold excess of unlabeled FAP8 base ligand at 4 hours post injection (Fig.39B). [0375] In a third study, human breast cancer (MDA-MB-231) tumor-bearing mice were intravenously injected with either (a) [ 111 In] In-FAP8-PEG3-IP-DOTA (In-Conjugate 24') (17.2 MBq/cc) alone (Fig. 40A); or (b) In-Conjugate 24' (17.2 MBq/cc) with unlabeled FAP8 base ligand for competition (Fig.40B). At indicated time points (1 hour, 4 hours, 24 hours, 72 hours, 120 hours, and 168 hours post-injection), mice were assessed and/or euthanized by CO 2 asphyxiation and organs of interest were harvested, excess blood was rinsed, dried, weighed, and then analyzed by gamma counter, and the results were decay corrected and presented as the percentage of the injected dose per gram of tissue (%ID/g) ± standard deviation (SD). In- Conjugate 24' in the MDA-MB-231 tumor bearing mice had 100-fold excess of unlabeled FAP8 base ligand at 4 hours post injection (Fig.40B). [0376] Still further, KB tumor-bearing mice were intravenously injected with either (a) [ 111 In] In- FAP8-PEG 3 -IP-DOTA (In-Conjugate 24') (2.7 MBq/cc) alone (Fig.41A); or (b) In-Conjugate 24' (2.7 MBq/cc) with unlabeled FAP8 base ligand for competition (Fig. 41B). At indicated time points (1 hour, 4 hours, 24 hours, 72 hours, 120 hours, and 168 hours post-injection), mice were assessed and/or euthanized by CO2 asphyxiation and organs of interest were harvested, excess blood was rinsed, dried, weighed, and then analyzed by gamma counter, and the results were decay corrected and presented as the percentage of the injected dose per gram of tissue (%ID/g) ± standard deviation (SD). In-Conjugate 24' in the KB tumor bearing mice had 100-fold excess of unlabeled FAP8 base ligand at 4 hours post injection (Fig.41B). [0377] Example 16 Radiolabelling and Efficacy Studies [0378] FAP8-PEG3-IP-DOTA (Conjugate 24') was dissolved in ammonium acetate buffer (1.0 M, pH 7.0) and labeled with 111 In [InCl 3 ] (BWMX Canada Ltd., Cambridge, Ontario, CA) or 177 Lu [LuCl3] (RadioMedix, Inc., Houston, TX) to obtain a specific activity of ≤ 3 MBq/nmol or ≤7.50 MBq/nmol, respectively. The resulting solutions were heated at 90 ºC for 5 minutes and the radiopurities of the products were analyzed by radio-HPLC. Radio purity exceeded 95% in all studies. [0379] Efficacy studies were conducted on mice expressing tumors from various cell lines. Briefly, Balb/c mice were inoculated on their shoulders with 1 x 10 5 cells of 4T1 cells, whereas nu/nu mice were inoculated on their shoulders with 5 x 10 6 cells of HT29 (a human colorectal adenocarcinoma cell line), KB (tumor cell line HeLa), MDA-MB-231 (a human breast cancer cell line), or HEK-FAP cells. [0380] 4T1, HT29, KB, and MDA-MB231 tumor-bearing mice were randomly divided into control and treatment groups (n = 5/group) to ensure similar starting tumor volumes. Each cohort received a single intravenous injection of saline or with 37 MBq/mouse of [ 177 Lu] Lu-FAP8- PEG3-IP-DOTA conjugate (Lu-Conjugate 24') to HT29, KB and MDA-MB231 tumor-bearing mice and 37 MBq, 18.5 MBq and 9.25 MBq/mouse of [ 177 Lu] Lu-FAP8-PEG 3 -IP-DOTA (Lu- Conjugate 24') to 4T1 tumor bearing mice on day 0. [0381] Tumors were then measured with a caliper in two perpendicular directions every other day. Mice were euthanized upon reaching one of the predefined endpoint criteria according to IACUC regulations. [0382] Mice were weighed every other day during radiotherapy as a gross evaluation of health. Tissue sections from organs of interest (n = 1–8 per organ per mouse) were preserved and examined blindly for lesions by a board-certified veterinary pathologist. [0383] As shown in Fig.37, treatment with Lu-Conjugate 24' had a positive effect on both tumor growth and overall survival in all cell lines as compared to the control groups. Further, the treatment did not adversely affect the subject’s weight. [0384] Additionally, different doses of [ 177 Lu] Lu-FAP8-IP-DOTA (Lu-Conjugate 24') were also assessed. Briefly, Balb/c mice were inoculated on their shoulders with 1 x 10 5 cells of 4T1 cells. Thereafter, treatment groups were treated with 37.0 MBq, 18.5 MBq, or 9.25 mBq of [ 177 Lu] Lu- FAP8-IP-DOTA (Lu-Conjugate 24'). Control groups (n = 5) were treated with PBS (Fig.38). Example 17 Radio-HPLC Chromatogram of FAP8-NOTA (Conjugate 18') Chelation with 64 CuCl 2 [0385] A mixture of radioactivity ( 64 CuCl2, 8.5 mCi, 30 µL) + FAP8-PEG3-NOTA (Conjugate 18') (8.5 µL of 5mM stock) + in 100 µL of NH 4 OAc (pH = 7, 1M) was incubated at room temperature for 20 minutes. The progress of the reaction was monitored by radio HPLC. The reaction mixture was further diluted with 5% EtOH in PBS and used for binding affinity studies and injections conducted using the protocols described above. As shown in Fig. 42, 100% chelation was achieved after 20 minutes, with stability at both 2 and 6 hours. [0386] Mice were inoculated with U87Mg cells subcutaneously using known protocols. When tumors grew to about 200-300 mm 3 , the mice were treated with an injection Conjugate 18' chelated with 64 CuCl2 (5 nmol/mouse) or a competition ligand, followed by a radioactivity dose of 1 mCi/mouse. The mice were monitored over time and imaged to visualize radioactivity and conjugate uptake. [0387] As shown in Fig. 43, there was a significant tumor uptake of the 64 Cu-Conjugate 18' between 4 to 24 hours post treatment. In particular, there was specific tumor uptake (white arrows indicating tumor). Example 18 Dose Escalation Analyses [0388] Dose escalation analyses of [ 111 In] In-FAP8-PEG 3 -IP-DOTA (In-Conjugate 24') in 4T1 tumor-bearing mice was performed (n = 3) with 1.0 nmol/mouse, 2.5 nmol/mouse, 5.0 nmol/mouse, or 10.0 nmol/mouse doses of Conjugate 24' provided with constant radioactivity (0.74 MBq/mouse) and measured at three different times (4 hours, 24 hours, and 120 hours post- injection). Fig.44 shows the percent injected dose/gram of tissue and in vivo distribution measured at the different time points. [0389] Dose escalation analyses of [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') in 4T1 tumor-bearing mice was performed (n = 3) with 0.3 nmol/mouse (Fig. 45A) or 1.0 nmol/mouse (Fig. 45B) of Lu-Conjugate 24' provided with constant radioactivity (0.74 MBq/mouse) and measured at different times (1 hour, 4 hours, 24 hours, 72 hours, 120 hours, and 168 hours post- injection). [0390] Comparisons of the total absorbed dose efficient in selected organs (Blood, Heart, Lungs, Liver, Spleen, Kidneys, and Bone Marrow, and in the tumors of 4T1 tumor-bearing mice) are shown in Figs.46A-46D. Example 19 Toxicology Studies [0391] To assess toxicology in the HT29 tumor-bearing mice treated with Lu-Conjugate 24', 4 µm sections of mouse heart, liver, and kidney tissue samples were taken following radiotherapy treatments of a single dose of [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') and 37 MBq radiotherapy in athymic nu/nu mice bearing HT29 tumors. 1.0 mCi was used for the treatment. The tissue samples were stained with H&E and representative photomicrographs were taken (Fig. 47; Table 13). Table 13. 177 Lu-FAP8-PEG 3 -IP-DOTA (Lu-Conjugate 24') Treatment on HT29 Tumor Bearing Mice N o d ag ostc es o s [0392] The X identified in Table 13 in connection with the liver relates to a multifocally, hepatocellular dropout that was identified with associated mild inflammation. These foci were uncommon, but not associated with typically locations of extramedullary haematopoiesis. Mononuclear cells were present around portal areas multifocally. Hepatic extramedullary haematopoiesis was present and expected in the mice. [0393] The X identified in Table 13 in connection with the heart relates to a focal area of epicardium at the apex of the heart that contained a small number of mononuclear infiltrates and hypertrophied epicardial mesothelial cells. The lesion was mild and the significance unknown. Evidence of infection was not present. [0394] The X identified in Table 13 in connection with the kidneys relates to diffuse, morphologic changes in the kidneys that were consistent with chronic progressive nephropathy, which is a spontaneous lesion in rats and mice. Diagnostic features observed in this mouse were thickened tubular basement membranes and tubular epithelial nuclear crowding and basophilia. Hyaline casts were present in medullary tubules. Multifocal glomerular atrophy and sclerosis and mononuclear cell infiltration were observed. Miled interstitial fibrosis was observed. [0395] Overall, there was no significant difference in histopathologic findings between treated and untreated mice in this study. [0396] To assess toxicology in the MDA-MB-231 tumor-bearing mice treated with Lu-Conjugate 24', 4 µm sections of mouse heart, liver, and kidney tissue samples were taken following radiotherapy treatments of a single dose of [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') and 37 MBq radiotherapy in athymic nu/nu mice bearing MDA-MB-231 tumors. 1.0 mCi was used for the treatment. The tissue samples were stained with H&E and representative photomicrographs were taken (Fig.48; Table 14). Table 14. 177 Lu-FAP8-PEG 3 -IP-DOTA (Lu-Conjugate 24') Treatment on MDA-MB-231 Tumor Bearing Mice g [0397] The X related to M8 identified in Table 14 in connection with the liver relates to the liver having one small focus of hepatocellular dropout with a few granulocytes. The lesion was mild and unlikely to affect the animal clinically. The X related to M9 identified in Table 14 in connection with the liver relates to the liver having multifocal random areas of inflammation containing neutrophils and macrophages associated with hepatocellular necrosis. [0398] 2 of the 5 mice (#8 and #9) in this treated group had histopathologic lesions consistent with necrosis and active inflammation. The non-treated mice did not have lesions. [0399] To assess toxicology in the KB tumor-bearing mice treated with Lu-Conjugate 24', 4 µm sections of mouse heart, liver, and kidney tissue samples were taken following radiotherapy treatments of a single dose of [ 177 Lu] Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') and 37 MBq radiotherapy in athymic nu/nu mice bearing KB tumors.1.0 mCi was used for the treatment. The tissue samples were stained with H&E and representative photomicrographs were taken (Fig.49; Table 15). Table 15. 177 Lu-FAP8-PEG3-IP-DOTA (Lu-Conjugate 24') Treatment on KB Tumor Bearing Mice [0400] The X related identified in Table 15 in connection with the kidneys relates to a focal area of mononuclear inflammation in the renal interstitium. The lesion was mild and not likely clinically relevant. [0401] The X related to M4 identified in Table 15 in connection with the liver relates to mononuclear cells that infiltrated and surrounded the periportal areas multifocally. Rarely, there can be random areas of hepatocellular drop out and mononuclear cell infiltrates. [0402] The X related to M 6 identified in Table 15 in connection with the liver relates to mononuclear cells that infiltrated and surrounded the periportal areas multifocally. [0403] 2 of the 3 mice (#4 and #6) in this KB treated group had histopathologic lesions consistent with mononuclear inflammation around portal areas. The inflammation did not contain necrosis or neutrophil leukocytes as that seen in the MDA-MB-231 group. The non-treated mice did not have lesions. Example 20 FAP8-PEG3-S0456 and FAP8-PEG3-IR-Dye800CW Conjugates [0404] Using the protocols described herein in connection with the above Examples, FAP8-PEG 3 - IR-Dye800CW was administered to 4T1 tumor bearing mice and whole body and ex vivo fluorescence imaging performed at different time points (Fig.50). A competition study was also performed as previously described, with FAP8-PEG3-IR-Dye800CW (Conjugate 58a) administered to 4T1 tumor-bearing mice and imaged at 6 hours post injection (Fig.51). [0405] Dose escalation studies were also conducted on FAP8-PEG3-S0456 in KB tumor bearing mice. The FAP8-PEG 3 -S0456 conjugate was administered at different concentrations (10 nmol, 5 nmol, 2.5 nmol and 1.25 nmol) to the test groups, and the mice were whole body and ex vivo imaged at 4 hours post injection (Fig. 52). The organs shown in Fig. 52 (from top to bottom) include tumor, heart, lung, liver, spleen, stomach, intestine, kidneys, and muscle. [0406] The same study was repeated using 5 nmol of FAP8-PEG3-S0456 in KB tumor bearing mice (Fig.53), 5 nmol of FAP8-PEG3-S0456 in HT29 tumor bearing mice (Fig.54), and 5 nmol of FAP8-PEG3-S0456 in U87Mg tumor bearing mice (Fig.55), with each image taken at 4 hours post injection. [0407] Additionally, images were taken of 4T1 tumor bearing mice at 4 hours and 12 hours post injection (5 nmol) of FAP8-PEG 3 -S0456 (Fig.56), and of 4T1 tumor-bearing mice at 4 hours post injection (5 nmol) of either FAP8-PEG6-S0456 (Fig.57) or FAP8-PEG8-S0456 (Fig.58).