Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PSMA TARGETED CONJUGATE COMPOUNDS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/183633
Kind Code:
A1
Abstract:
PSMA targeted conjugate compounds, pharmaceutical compositions comprising these compounds, methods for treating and detecting cancers in a subject, methods for identifying cancer cells in a sample are described herein.

Inventors:
BASILION JAMES P (US)
WANG XINNING (US)
WALKER NATALIE (US)
Application Number:
PCT/US2019/023901
Publication Date:
September 26, 2019
Filing Date:
March 25, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CASE WESTERN RESERVE UNIVERISTY (US)
International Classes:
A61K47/18; A61K47/65; A61N5/01
Domestic Patent References:
WO2014127365A12014-08-21
Foreign References:
US20150366968A12015-12-24
US20160067341A12016-03-10
Attorney, Agent or Firm:
SUTKUS, Richard A. (US)
Download PDF:
Claims:
Having described the invention, we claim:

1. A compound comprising the general formula (I):

wherein n and n1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

m at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

2. The compound of claim 1, wherein Y and Y1 are each independently selected from the group consisting of an imaging agent, anticancer agent, and combinations thereof.

3. The compound of claim 1, wherein L includes at least one ring selected from the group consisting of an optionally substituted 4 to 7 membered nonaromatic heterocyclic ring and an optionally substituted C4-C7 cycloalkyl ring.

4. The compound of claim 1, wherein B has the following formula:

wherein m is 1, 2, 3, or 4; and Y and Y1 are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

5. The compound of claim 4, wherein Y is a therapeutic agent and Y1 is a detectable moiety.

6. The compound of claim 1, wherein the therapeutic agent is linked to B via a protease cleavable or acid-labile linker.

7. The compound of claim 6, wherein the linker is selected from a MC-Val-Cit- PABC protease cleavable linker and a 4-(4-maleimidomethyl)cyclohexane-l-carboxyl hydrazide (MMCCH) acid-labile linker.

8. The compound of claim 7, wherein the therapeutic agent is selected from doxorubicin and monomethyl auristatin E (MMAE).

9. The compound of claim 1, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; A is H or

are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

10. The compound of claim 9, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; and Y and Y1 are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

11. The compound of claim 9, the compound having a formula selected from the group consisting of:

12. A composition for diagnosing and/or treating cancer, the composition comprising a prostate specific membrane antigen (PSMA) targeted conjugate compound that includes general formula (I):

wherein n and n1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

m at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

13. The composition of claim 12, wherein Y and Y1 are each independently selected from at least one of an imaging agent, anticancer agent, or combination thereof.

14. The composition of claim 12, wherein L includes at least one ring selected from the group consisting of an optionally substituted 4 to 7 membered nonaromatic heterocyclic ring and an optionally substituted C4-C7 cycloalkyl ring.

15. The composition of claim 12, wherein B has the following formula:

wherein m is 1, 2, 3, or 4; and Y and Y1 are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

16. The composition of claim 15, wherein Y is a therapeutic agent and Y1 is a detectable moiety.

17. The composition of claim 12, wherein Y is therapeutic agent linked to B via a protease cleavable or acid-labile linker.

18. The composition of claim 17, wherein the linker is selected from a MC-Val- Cit-PABC protease cleavable linker and a 4-(4-maleimidomethyl)cyclohexane-l -carboxyl hydrazide (MMCCH) acid-labile linker.

19. The composition of claim 18, wherein the therapeutic agent is selected from doxorubicin and monomethyl auristatin E (MMAE).

20. The composition of claim 12, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; A is H or

are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

21. The composition of claim 20, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; and Y and Y1 are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

22. The composition of claim 20, the PSMA targeted conjugate compound having a formula selected from the group consisting of:

pharmaceutical salts thereof.

23. A composition for treating prostate cancer in a subject, the composition comprising a PSMA targeted conjugate compound having the general formula:

wherein n and n1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group;

B has the following formula:

wherein Y is an anticancer agent directly or indirectly linked to B; and Y1 is selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to A.

24. The composition of claim 23, wherein Y1 is selected from at least one of an imaging agent, anticancer agent, or combination thereof.

25. The composition of claim 23, wherein L includes at least one ring selected from the group consisting of an optionally substituted 4 to 7 membered nonaromatic heterocyclic ring and an optionally substituted C4-C7 cycloalkyl ring.

26. The composition of claim 23, wherein B has the following formula:

wherein m is 1, 2, 3, or 4; wherein Y is an anticancer agent and Y1 is a detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

27. The composition of claim 23, wherein Y is linked to B via a protease cleavable or acid-labile linker.

28. The composition of claim 27, wherein the linker is selected from a MC-Val- Cit-PABC protease cleavable linker and a 4-(4-maleimidomethyl)cyclohexane-l -carboxyl hydrazide (MMCCH) acid-labile linker.

29. The composition of claim 28, wherein the anticancer agent is selected from doxorubicin and monomethyl auristatin E (MMAE).

30. The composition of claim 23, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; wherein Y is an

anticancer agent; detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

31. The composition of claim 30, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; wherein Y is an anticancer agent and Y1 is a detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

32. The composition of claim 30, the PSMA targeted conjugate compound having a formula selected from the group consisting of:

pharmaceutical salts thereof.

33. A method for treating cancer in a subject comprising:

administering to the subject a therapeutically effective amount of a composition comprising a PSMA targeted conjugate compound having the general formula:

wherein n and n1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group;

B has the following formula:

wherein Y is an anticancer agent directly or indirectly linked to B; and Y1 is selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to A.

34. The method of claim 33, wherein the composition is administered

systemically.

35. The method of claim 34, wherein the composition is administered by intravenous injection.

36. The method of claim 33, wherein Y1 is selected from at least one of an imaging agent, anticancer agent, or combination thereof.

37. The method of claim 33, wherein L includes at least one ring selected from the group consisting of an optionally substituted 4 to 7 membered nonaromatic heterocyclic ring and an optionally substituted C4-C7 cycloalkyl ring.

38. The method of claim 33, wherein B has the following formula:

wherein m is 1, 2, 3, or 4; wherein Y is an anticancer agent and Y1 is a detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

39. The method of claim 33, wherein Y is linked to B via a protease cleavable or acid-labile linker.

40. The method of claim 39, wherein the linker is selected from a MC-Val-Cit- PABC protease cleavable linker and a 4-(4-maleimidomethyl)cyclohexane-l-carboxyl hydrazide (MMCCH) acid-labile linker.

41. The method of claim 40, wherein the anticancer agent is selected from doxorubicin and monomethyl auristatin E (MMAE).

42. The method of claim 33, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; wherein Y is an

anticancer agent; detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

43. The method of claim 42, comprising the general formula:

wherein m, n and n1 are each independently 1, 2, 3, or 4; wherein Y is an anticancer agent and Y1 is a detectable moiety; and wherein Y and Y1 are directly or indirectly linked to B.

44. The method of claim 42, the PSMA targeted conjugate compound having a formula selected from the group consisting of:

pharmaceutical salts thereof.

45. The method of claim 33, wherein the cancer is a PSMA expressing cancer.

46. The method of claim 45, wherein the cancer is prostate cancer.

Description:
PSMA TARGETED CONJUGATE COMPOUNDS AND USES THEREOF

RELATED APPLICATION

[0001] This application claims priority from U.S. Provisional Application

No. 62/647,267, filed March 23, 2018, the subject matter of which is incorporated herein by reference in its entirety.

TECHNICAL FIELD

[0002] This application relates to prostate-specific membrane antigen (PSMA) targeted conjugate compounds and to their use in compositions for targeting, imaging, and treating cancer.

BACKGROUND

[0003] Prostate-specific membrane antigen (PSMA) is a 120 kDa protein expressed in prostate tissues and was originally identified by reactivity with a monoclonal antibody designated 7E11-C5 (Horoszewicz et al., 1987, Anticancer Res. 7:927-935; U.S. Patent No. 5,162,504). PSMA is characterized as a type II transmembrane protein sharing sequence identity with the transferrin receptor (Israeli et al., 1994, Cancer Res. 54:1807-1811). PSMA is a glutamate carboxy-peptidase that cleaves terminal carboxy glutamates from both the neuronal dipeptide N-acetylaspartylglutamate (NAAG) and gamma-linked folate

polyglutamate. That is, expression of PSMA cDNA confers the activity of N-acetylated a- linked acidic dipeptidase or "NAALADase" activity (Carter et al., 1996, PNAS 93:749-753).

[0004] PSMA is expressed in increased amounts in prostate cancer, and elevated levels of PSMA are detectable in the sera of these patients (Horoszewicz et al., 1987, supra; Rochon et al., 1994, Prostate 25:219-223; Murphy et al., 1995, Prostate 26:164-168; and Murphy et al., 1995, Anticancer Res. 15:1473-1479). As a prostate carcinoma marker, PSMA is believed to serve as a target for imaging and cytotoxic treatment modalities for prostate cancer.

Prostate carcinogenesis, for example, is associated with an elevation in PSMA abundance and enzymatic activity of PSMA. PSMA antibodies, particularly indium-l ll labeled and tritium labeled PSMA antibodies, have been described and examined clinically for the diagnosis and treatment of prostate cancer. PSMA is expressed in prostatic ductal epithelium and is present in seminal plasma, prostatic fluid and urine.

[0005] Recent evidence suggests that PSMA is also expressed in tumor associated neovasculature of a wide spectrum of malignant neoplasms including conventional (clear cell) renal carcinoma, transitional cell carcinoma of the urinary bladder, testicular embryonal carcinoma, colonic adenocarcinoma, neuroendocrine carcinoma, gliobastoma multiforme, malignant melanoma, pancreatic ductal carcinoma, non-small cell lung carcinoma, soft tissue carcinoma, breast carcinoma, and prostatic adenocarcinoma. (Chang et al. (1999) Cancer Res. 59, 3192-3198).

SUMMARY

[0006] Embodiments described herein relate to PSMA targeted conjugate compounds, pharmaceutical compositions comprising these compounds, methods for treating and detecting cancers (e.g., prostate cancer) in a subject using these PSMA targeted conjugate compounds, and methods for identifying cancer cells (e.g., prostate cancer cells) in a sample using these compounds.

[0007] In some embodiments, the compound can include the general formula (I):

wherein n and n 1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group;

B has the following formula:

; wherein m is 1, 2, 3, or 4, A is H or are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

[0008] Other embodiments relate to a composition for diagnosing and/or treating cancer, the composition comprising a prostate specific membrane antigen (PSMA) targeted conjugate compound that includes general formula (I):

wherein n and n 1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

wherein m is 1, 2, 3, or 4, A is H or

are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B.

[0009] Another embodiment relates to a composition for treating prostate cancer in a subject, the composition comprising a PSMA targeted conjugate compound having the general formula:

wherein n and n 1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

wherein m is 1, 2, 3, or 4, A is H or

in Y is an anticancer agent directly or indirectly linked to B; and Y 1 is selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to A.

[0010] Still other embodiments relate to a method of treating cancer, such as prostate cancer, in a subject. The method includes administering to the subject a therapeutically effective amount of a composition that includes a prostate specific membrane antigen (PSMA) targeted conjugate compound having the general formula (I):

wherein n and n 1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

wherein m is 1, 2, 3, or 4, A is H or

wherein Y is an anticancer agent directly or indirectly linked to B; and Y 1 is selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to A. The anti-cancer agent can be selected from doxorubicin or monomethylauristatin E (MMAE).

BRIEF DESCRIPTION OF THE DRAWINGS

[0011] The following is a brief description of the drawings which are presented for the purpose of illustrating the invention and not for the purpose of limiting them.

[0012] Fig. 1 illustrates the outline of Structures of Three PSMA-l -Doxorubicin Prodrug Conjugates.

[0013] Fig. 2 illustrates proposed mechanisms for prodrug release of free doxorubicin.

[0014] Fig. 3 illustrates a structure of PSMA-l(Cys)

[0015] Fig. 4 illustrates the synthesis of SMCC-Dox

[0016] Fig. 5 illustrates the synthesis of Non-cleavable PSMA-l-SMCC-Dox.

[0017] Fig. 6 illustrates the synthesis of MC-Val-Cit-PABC-Dox

[0018] Fig. 7 illustrates the synthesis of cathepsin-cleavable PSMA-l -MC-Val-Cit-

PABC-Dox

[0019] Fig. 8 illustrates the synthesis of MMCCH-Dox

[0020] Fig. 9 illustrates synthesis of acid-labile PSMA-l -MMCCH-Dox.

[0021] Fig. 10 illustrates absorption spectra of free doxorubicin and PSMA-Targeted conjugates. Note, absorption spectra was measured from stock dilutions and concentration is not uniform. While the absorbance values are not identical for this reason, the shape of the curve and peak absorption wavelength remains unchanged by conjugation to PSMA-l (Cys). For brevity, PSMA-l-SMCC-Dox is labeled PSD, PSMA-l-MC-Val-Cit-PABC-Dox is labeled PCD, and PSMA-l-MMCCH-Dox is labeled PMD in this figure.

[0022] Figs. 1 l(A-B) illustrate in vitro drug uptake and cellular localization by fluorescence microscopy. PSMA(+) PC3Pip cells and PSMA(-) PC3Flu cells were treated with free doxorubicin or one of three PSMA-targeted conjugates. DAPI nuclear staining is shown in blue, and doxorubicin fluorescence signal is seen in red. Specific uptake of PSMA- l-Doxorubicin conjugates is seen in PC3Pip cells, more so at short incubation periods, A.)l mM, 90 minutes, compared to long periods, B.) 1 pM, 48 hours. Only free doxorubicin and PSMA-l-MMCCH-Dox showed colocalization of the red doxorubicin and blue DAPI nuclear staining. Note negligible autofluoresnce in the red channel for PC3Flu cells, seen in the untreated control slide.

[0023] Figs. 12(A-B) illustrate 48 Hour Cytotoxicity of Free and PSMA-Targeted Doxorubicin. PSMA(+) PC3Pip cells and PSMA(-)PC3Flu cells were treated with doxorubicin or PSMA-l -Doxorubicin conjugates for 48 hours before measuring cell viability relative to untreated controls using Dojindo CCK8 assays. Doses in nM. Same data displayed both by drug (A) and as a single graph with all drugs overlay ed (B) for easier visual comparison. For brevity, PSMA-l-SMCC-Dox is labeled PSD, PSMA-l -MC-Val-Cit- PABC-Dox is labeled PCD, and PSMA-l-MMCCH-Dox is labeled PMD in this figure. Note maximum dose tested for PMD was 2000 nM instead of 4000 nM for the other drugs. Stats performed on variation within six wells tested.

[0024] Figs. 13(A-B) illustrate cytotoxicity of Dox and PSMA-MMCCH-Dox,

Alternate Conditions. PSMA(+) PC3Pip cells and PSMA(-) PC3Flu cells were treated with free doxorubucin and PSMA-l -MMCH-Dox for 24 hours, followed by 48 hours of growth in fresh media and determination of cell viability with Dojindo CK8 Assay. Doses in nM. A.) Cell viability data, showing decreased viability of PC3Pip cells compared to PC3Flu cells when treated with PSMA-l-MMCCH-Dox B.) Cell viability after treatment with PSMA-l- MMCCH-Dox relative to cell viability after treatment with free doxorubicin, generally demonstrating decreased efficacy of PSMA-l-MMCCH-Dox in culture for both cell lines. Asterisks represent a statistically significant difference from a ratio of 1.

[0025] Fig. 14 (A-B) illustrate tumor response and mouse weight. A.) Tumor growth was monitored by volume using calipers and is expressed here as a ratio of initial Day 0 volume. Treatment days indicated with vertical arrows. While all tumors increased in size, tumors of mice treated with PMD (PSMA-l-MMCCH-Dox) grew more slowly than mice treated with free doxorubicin. N=3 per group. B.) Mice treated with PMD experienced less weight loss by the end of the 21 day trial period.

[0026] Fig. 15 illustrates a graph showing competition binding studies of PSMA- MMAE-Cy5.5.

[0027] Fig. 16 illustrates a graph showing in vitro cytotoxicity experiments of PSMA- MMAE-Cy5.5. PSMA-MMAE-Cy5.5 is more effective in killing PC3pip cells.

[0028] Fig. 17 illustrates in vivo images of PSMA-MMAE-Cy5.5 showing selective binding was observed in PC3pip tumors.

[0029] Fig. 18 illustrates ex vivo images of organs at 72-hour post-injection of PSMA- MMAE-Cy5.5. Signals were only observed in PC3pip tumor and kidneys.

[0030] Fig. 19 illustrates graph showing a comparison of fluorescent signal intensity on PC3pip and PC3fly tumor. Signal peaked at 24-hour post-injection of PSMA-MMAE-Cy5.5. At this time point, the fluorescent intensity in PC3pip tumor is about 4-fold higher than that in PC3flu tumors.

[0031] Fig. 20 illustrates in vitro images of PC3 pip and PC3flu cells incubated 4 hours with 50nm PSMA-MMAE-Cy5.5 with and without lOOx ZJ24, a small molecule targeting prostate specific membrane antigen (PSMA).

[0032] Fig. 21 illustrates a graph showing the cytotoxicity of PSMA-MMAE using 1,000 newly sorted PC3pip cells following a 72-hour incubation. PSMA-MMAE-Cy5.5 is about l7-fold more potent to PC3pip cells than to PC3flu cells.

DETAILED DESCRIPTION

[0033] All scientific and technical terms used in this application have meanings commonly used in the art unless otherwise specified. The definitions provided herein are to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the application.

[0034] The articles "a" and "an" are used herein to refer to one or to more than one (/. <? ., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.

[0035] As used herein, the term“sample” can refer to a specimen or culture obtained from any source, as well as clinical, research, biological and environmental samples. Biological samples may be obtained from animals (including humans) and encompass cells, fluids, solids, tissues, and organs, and whole organisms.

[0036] As used herein, the term“subject” can refer to any animal including, but not limited to, humans and non-human animals (e.g., rodents, arthropods, insects, fish

(e.g., zebrafish)), non-human primates, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, or canines felines, aves, etc.).

[0037] As used herein, the terms“cancer” or“tumor” refer to any neoplastic growth in a subject, including an initial tumor and any metastases. The cancer can be of the liquid or solid tumor type. Liquid tumors include tumors of hematological origin, including, e.g., myelomas (e.g., multiple myeloma), leukemias (e.g., Waldenstrom's syndrome, chronic lymphocytic leukemia, other leukemias), and lymphomas (e.g., B-cell lymphomas, non- Hodgkin’s lymphoma). Solid tumors can originate in organs and include cancers of the lungs, brain, breasts, prostate, ovaries, colon, kidneys and liver.

[0038] As used herein, the terms“cancer cell” or“tumor cell” can refer to cells that divide at an abnormal (i.e., increased) rate. Cancer cells include, but are not limited to, carcinomas, such as squamous cell carcinoma, non-small cell carcinoma (e.g., non-small cell lung carcinoma), small cell carcinoma (e.g., small cell lung carcinoma), basal cell carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, adenocarcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, undifferentiated carcinoma, bronchogenic carcinoma, melanoma, renal cell carcinoma, hepatoma-liver cell carcinoma, bile duct carcinoma, cholangiocarcinoma, papillary carcinoma, transitional cell carcinoma, choriocarcinoma, semonoma, embryonal carcinoma, mammary carcinomas, gastrointestinal carcinoma, colonic carcinomas, bladder carcinoma, prostate carcinoma, and squamous cell carcinoma of the neck and head region; sarcomas, such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordosarcoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, synoviosarcoma and

mesotheliosarcoma; hematologic cancers, such as myelomas, leukemias (e.g., acute myelogenous leukemia, chronic lymphocytic leukemia, granulocytic leukemia, monocytic leukemia, lymphocytic leukemia), lymphomas (e.g., follicular lymphoma, mantle cell lymphoma, diffuse large B-cell lymphoma, malignant lymphoma, plasmocytoma, reticulum cell sarcoma, or Hodgkin’s disease), and tumors of the nervous system including glioma, glioblastoma multiform, meningoma, medulloblastoma, schwannoma and epidymoma. [0039] As used herein, the term“polypeptide” can refer to an oligopeptide, peptide, polypeptide, or protein sequence, or to a fragment, portion, or subunit of any of these, and to naturally occurring or synthetic molecules. The term“polypeptide” can also include amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain any type of modified amino acids. The term can also include peptides and polypeptide fragments, motifs and the like, glycosylated polypeptides, and all“mimetic” and “peptidomimetic” polypeptide forms.

[0040] PSMA" refers to Prostate Specific Membrane Antigen, a potential carcinoma marker that has been hypothesized to serve as a target for imaging and cytotoxic treatment modalities for cancer.

[0041] As used herein, the term“small molecule” can refer to lipids, carbohydrates, polynucleotides, polypeptides, or any other organic or inorganic molecules.

[0042] As used herein, the term“imaging probe” can refer to a biological or chemical moiety that may be used to detect, image, and/or monitor the presence and/or progression of a cell cycle, cell function/physiology, condition, pathological disorder and/or disease.

[0043] As used herein, the terms“treating” or“treatment” of a disease can refer to executing a treatment protocol to eradicate at least one diseased cell. Thus,“treating” or “treatment” does not require complete eradication of diseased cells.

[0044] As used herein, the term "nanoparticle" refers to any particle having a diameter of less than 1000 nanometers (nm). In some embodiments, nanoparticles can be optically or magnetically detectable. In some embodiments, intrinsically fluorescent or luminescent nanoparticles, nanoparticles that comprise fluorescent or luminescent moieties, plasmon resonant nanoparticles, and magnetic nanoparticles are among the detectable nanoparticles that are used in various embodiments. In general, the nanoparticles should have dimensions small enough to allow their uptake by eukaryotic cells. Typically, the nanoparticles have a longest straight dimension (e.g., diameter) of 200 nm or less. In some embodiments, the nanoparticles have a diameter of 100 nm or less. Smaller nanoparticles, e.g., having diameters of 50 nm or less, e.g., about 1 nm to about 30 nm or about 1 nm to about 5 nm, are used in some embodiments.

[0045] As used herein, an“effective amount” can refer to that amount of a therapeutic agent that results in amelioration of symptoms or a prolongation of survival in the subject and relieves, to some extent, one or more symptoms of the disease or returns to normal (either partially or completely) one or more physiological or biochemical parameters associated with or causative of the disease.

[0046] As used herein, therapeutic agents can include any agent (e.g., molecule, drug, pharmaceutical composition, etc.) capable of preventing, inhibiting, or arresting the symptoms and/or progression of a disease.

[0047] Embodiments described herein relate to PSMA targeted conjugate compounds, as well as pharmaceutical compositions comprising these conjugate compounds, methods for treating and detecting cancers, such as prostate cancer, in a subject using these compounds, and methods for identifying cancer cells in a sample using these compounds.

[0048] Using a modified PSMA-l peptide ligand, PSMA ligands (PSMA-l(Cys)) have been developed that include a terminal cysteine residue that allows for coupling of the thiol group to maleimide linkers and/or the cysteine residue amine group to a carbon linker to create PSMA targeted compounds conjugated to one or more detectable moiety, therapeutic agent, or a theranostic agent. It has been shown that PSMA targeted compounds conjugated to anti-cancer agents can increase uptake of the conjugate compound in PSMA expressing cells while also improving cell killing compared to agents administered alone. In addition, PSMA targeted conjugate compounds described herein can decrease off-target toxicity of the therapeutic agent, or a theranostic agent administered (e.g., systemically) to a subject.

[0049] Pathological studies indicate that PSMA is expressed by virtually all prostate cancers, and its expression is further increased in poorly differentiated, metastatic, and hormone-refractory carcinomas. Higher PSMA expression is also found in cancer cells from castration-resistant prostate cancer patients. Increased PSMA expression is reported to correlate with the risk of early prostate cancer recurrence after radical prostatectomy. In addition to being overexpressed in prostate cancer (PCa), PSMA is also expressed in the neovasculature of neoplasms including but not limited to conventional (clear cell) renal carcinoma, transitional cell carcinoma of the urinary bladder, testicular embryonal carcinoma, colonic adenocarcinoma, neuroendocrine carcinoma, gliobastoma multiforme, malignant melanoma, pancreatic ductal carcinoma, non-small cell lung carcinoma, soft tissue carcinoma, breast carcinoma, and prostatic adenocarcinoma.

[0050] In some embodiments, the PSMA targeted conjugate compounds described herein, can selectively recognize PSMA-expressing tumors, cancer cells, and/or cancer neovasculature in vivo and be used to deliver a therapeutic agent, detectable moiety, and/or theranostic agent to the PSMA-expressing tumors, cancer cells, and/or cancer neovasculature to treat and/or detect the PSMA-expressing tumors, cancer cells, and/or cancer

neovasculature in a subject.

[0051] In some embodiments, the PSMA expressing cancer that is treated and/or detected is prostate cancer. In other embodiments, the cancer that is treated and/or detected can include malignant neoplasms, such a conventional (clear cell) renal carcinoma, transitional cell carcinoma of the urinary bladder, testicular embryonal carcinoma, colonic adenocarcinoma, neuroendocrine carcinoma, gliobastoma multiforme, malignant melanoma, pancreatic ductal carcinoma, non-small cell lung carcinoma, soft tissue carcinoma, breast carcinoma, and prostatic adenocarcinoma.

[0052] In some embodiments, the compound can include a PSMA targeted conjugate compound that has the general formula (I):

wherein:

n and n 1 are each independently 1, 2, 3, or 4;

L is an optionally substituted aliphatic or heteroaliphatic linking group; B has the following formula:

, wherein m is 1, 2, 3, or 4, and A is H are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent that is directly or indirectly linked to B. In other embodiments, Y and Y 1 can each independently be selected from the group consisting of an imaging agent, anticancer agent, and combinations thereof.

[0053] In other embodiments, L can be an optionally substituted aliphatic or heteroaliphatic group that includes at least one ring selected from the group consisting of an optionally substituted 4 to 7 membered nonaromatic heterocyclic ring and an optionally substituted C4-C7 cycloalkyl ring.

[0054] An aliphatic group is a straight chained, branched or cyclic non-aromatic hydrocarbon, which is completely saturated or which contains one or more units of unsaturation. An alkyl group is a saturated aliphatic group. Typically, a straight chained or branched aliphatic group has from 1 to about 10 carbon atoms, preferably from 1 to about 4, and a cyclic aliphatic group has from 3 to about 10 carbon atoms, preferably from 3 to about 8. An aliphatic group is preferably a straight chained or branched alkyl group, e.g., methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl or octyl, or a cycloalkyl group with 3 to about 8 carbon atoms. C1-C4 straight chained or branched alkyl or alkoxy groups or a C3-C8 cyclic alkyl or alkoxy group (preferably C1-C4 straight chained or branched alkyl or alkoxy group) are also referred to as a "lower alkyl" or "lower alkoxy" groups; such groups substituted with -F, -Cl, -Br, or -I are "lower haloalkyl" or "lower haloalkoxy" groups; a "lower hydroxyalkyl" is a lower alkyl substituted with -OH; and the like.

[0055] Suitable optional substituents for a substitutable atom in alkyl, cycloalkyl, aliphatic, cycloaliphatic, heterocyclic, benzylic, aryl, or heteroaryl groups described herein are those substituents that do not substantially interfere with the activity of the disclosed compounds. A "substitutable atom" is an atom that has one or more valences or charges available to form one or more corresponding covalent or ionic bonds with a substituent. For example, a carbon atom with one valence available (e.g., -C(-H)=) can form a single bond to an alkyl group (e.g., -C(-alkyl)=), a carbon atom with two valences available (e.g., -C(H2)-) can form one or two single bonds to one or two substituents (e.g., -C(alkyl)(Br))-,

-C(alkyl)(H)-) or a double bond to one substituent (e.g., -C=0)-), and the like. Substitutions contemplated herein include only those substitutions that form stable compounds.

[0056] For example, suitable optional substituents for substitutable carbon atoms include -F, -Cl, -Br, -I, -CN, -N0 2 , -OR a , -C(0)R a , -0C(0)R a , -C(0)0R a , -SR a , -C(S)R a , -OC(S)R a , -C(S)OR a , -C(0)SR a , -C(S)SR a , -S(0)R a , -S0 2 R a , -S0 3 R a , -POR a R b , P0 2 R a R b , -P0 3 R a R b , -P0 4 R a R b , -P(S)R a R b , -P(S)OR a R b , -P(S)0 2 R a R b , -P(S)0 R a R b , -N(R a R b ), -C(0)N(R a R b ), -C(0)NR a NR b S0 2 R c , -C(0)NR a S0 2 R c , -C(0)NR a CN, -S0 2 N(R a R b ), -S0 2 N(R a R b ), -NR c C(0)R a , -NR c C(0)0R a , -NR c C(0)N(R a R b ), -C(NR c )-N(R a R b ), -NR d - C(NR c )-N(R a R b ), -NR a N(R a R b ), -CRC=CR a R b , -C=CR a , =0, =S, =CR a R b , =NR a , =NOR a , =NNR a , optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aliphatic, optionally substituted cycloaliphatic, optionally substituted heterocyclic, optionally substituted benzyl, optionally substituted aryl, and optionally substituted heteroaryl, wherein R a -R d are each independently -H or an optionally substituted aliphatic, optionally substituted cycloaliphatic, optionally substituted heterocyclic, optionally substituted benzyl, optionally substituted aryl, or optionally substituted heteroaryl, or, -N(R a R b ), taken together, is an optionally substituted heterocyclic group. Also contemplated are isomers of these groups.

[0057] Suitable substituents for nitrogen atoms having two covalent bonds to other atoms include, for example, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted aliphatic, optionally substituted cycloaliphatic, optionally substituted heterocyclic, optionally substituted benzyl, optionally substituted aryl, optionally substituted heteroaryl, -CN, -N0 2 , -OR a , -C(0)R a , -OC(0)R a , -C(0)OR a , -SR a , -S(0)R a , -S0 2 R a , -S0 3 R a , -N(R a R b ), -C(0)N(R a R b ), -C(0)NR a NR b S0 2 R c , -C(0)NR a S0 2 R c , -C(0)NR a CN, -S0 2 N(R a R b ), -S0 2 N(R a R b ), -NR c C(0)R a , -NR c C(0)OR a , -NR c C(0)N(R a R b ), and the like.

[0058] Suitable substituents for nitrogen atoms having three covalent bonds to other atoms include -OH, alkyl, and alkoxy (preferably C1-C4 alkyl and alkoxy). Substituted ring nitrogen atoms that have three covalent bonds to other ring atoms are positively charged, which is balanced by counteranions such as chloride, bromide, fluoride, iodide, formate, acetate and the like. Examples of other suitable counter anions are provided in the section below directed to suitable pharmacologically acceptable salts.

[0059] In some embodiments, B can have the following formula:

wherein m is 1, 2, 3, or 4 and Y can include at least one of a detectable moiety, therapeutic agent, or theranostic agent.

[0060] In some embodiments, B can have the following formula:

wherein m is 1, 2, 3, or 4 and Y and Y 1 are each independently selected from at least one of a detectable moiety, therapeutic agent, or theranostic agent.

[0061] In other embodiments, the compound can have the general formula:

wherein m, n and n 1 are each independently 1, 2, 3, or 4; A is H or

are each independently selected from at least one of a detectable moiety, therapeutic agent, or a theranostic agent.

[0062] In certain embodiments, the PSMA targeted conjugated compound can have the general formula:

wherein m, n, and n 1 are independently 1, 2, 3, or 4; and Y 2 is a H or can include at least one of an amino acid, peptide, detectable moiety, therapeutic agent, or theranostic agent.

[0063] In some embodiments, a therapeutic, imaging and/or a thernanostic agent can be coupled directly or indirectly to the PSMA targeted compound via a linker. The linker can include a heterobifunctional crosslinker capable of conjugating to the cysteine residue of the PSMA targeted compound via a maleimide group that is sulfhydryl (thiol; -SH) reactive.

[0064] In some embodiments, the heterobifunctional crosslinker can include a protease cleavable or acid-labile linker. In particular embodiments, the therapeutic, imaging and/or a theranostic agent can be coupled directly or indirectly to the PSMA targeted compound via an acid-labile linker, such as but not limited to 4-(4-maleimidomethyl)cyclohexane-l -carboxyl hydrazide (MMCCH) linkers where a hydrazine bond is cleaved to release a detectable moiety, therapeutic agent, or a theranostic agent coupled to the PSMA targeted compound.

In alternative embodiments, the therapeutic, imaging and/or a theranostic agent can be coupled directly or indirectly to the PSMA targeted portion of the compound ( PSMA- l(Cys)) via a non-cleavable linker, such as but not limited to Succinimidyl 4-(N- maleimidomethyl)cyclohexane- l-carboxylate (SMCC).

[0065] In other embodiments, the therapeutic, imaging and/or a theranostic agent can be coupled directly or indirectly to the PSMA targeted portion of the compound (PSMA-l(Cys)) via a protease cleavable linker. The protease cleavable linker can include a MC-Val-Cit- PABC protease cleavable linker where the Valine-Citrulline-PABC portion is the protease cleaveable linker.

[0066] In an exemplary embodiment, PSMA targeted conjugate compounds described herein are prepared by coupling a“vc-MMAE” linker-drug combination construct at the thiol group of the PSMA targeted compound cysteine residue. The vc-MMAE construct is also referred to as the linker-drug mc-vc-PABC-MMAE. The construct utilizes a

maleimidocaproyl (me) spacer, a protease- sensitive dipeptide, valine-citrulline (vc), a self- immolative spacer, para-amino benzyloxycarbonyl (PABC), and the antimitotic agent, monomethyl auristatin E (MMAE) The me spacer provides enough room for the vc group to be recognized by the lysosomal cysteine protease cathepsin B in a PSMA expressing cell, which cleaves the citrulline-PABC amide bond. The resultant PABC-substituted MMAE is not a stable intermediate and spontaneously undergoes a 1, 6-elimination with a loss of p- iminoquinone methide and carbon dioxide (self-immolation) leaving MMAE as the product to exert anti-mitotic effects.

[0067] In some embodiments, the PSMA targeted conjugate compound can include a detectable moiety directly or indirectly coupled to B at Y and/or Y 1 . Examples of detectable moieties include, but are not limited to: various ligands, radionuclides, fluorescent dyes, chemiluminescent agents, microparticles (such as, for example, quantum dots, nanocrystals, phosphors and the like), enzymes (such as, for example, those used in an ELISA, i.e., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase), colorimetric labels, magnetic labels, chelating groups, and biotin, dioxigenin or other haptens and proteins for which antisera or monoclonal antibodies are available.

[0068] In some embodiments, radionuclides can include atomic isotopes such as 99m Tc, min, 123 I, 124 I, 131 I, 67 Ga, 201 Tl, 125 I, 18 F, n C, 76 Br, 124 I, 68 Ga, 82 Rb, 13 N, 64 Cu, 90 Y, 188 Rh, T(tritium), 32 P, 35 S, 153 Sm, 89 Sr, 211 At, and 89 Zr. These isotopes can be directly or indirectly coupled to the PSMA ligand.

[0069] Fluorescence labeling agents or infrared labeling agents include those known to the art, many of which are commonly commercially available, such as ALEXA 350, 49-

PACIFIC BLUE, MARINA BLUE, ACRIDINE, EDANS, COUMARIN, BODIPY 493/503, CY2, BODIPY FL-X, DANSYL, ALEXA 488, FAM, OREGON GREEN, RHODAMINE GREEN-X, TET, ALEXA 430, CAL GOLD, BODIPY R6G-X, JOE, ALEXA 532, VIC, HEX, CAL ORANGE.TM., ALEXA 555, BODIPY 564/570, BODIPY TMR-X,

QUASAR.TM. 570, ALEXA 546, TAMRA, RHODAMINE RED-X, BODIPY 581/591, CY3.5, CY5.5, ROX, ALEXA 568, CAL RED, BODIPY TR-X, ALEXA 594, BODIPY 630/650-X, PULSAR 650, BODIPY 630/665-X, ALEXA 647, IR700, IR800, and QUASAR 670. Fluorescence labeling agents can include other known fluorophores, or proteins known to the art, for example, green fluorescent protein. The fluorescence labeling agents can be directly or indirectly coupled to the PSMA ligands, administered to a subject or a sample, and the subject/sample examined by fluorescence spectroscopy or imaging to detect the labeled compound. In some embodiments, the fluorescence labeling agent can include a cyanine monosuccinimidyl ester.

[0070] In an exemplary embodiment, the fluorescence labeling agent can include the cyanine monosuccinimidyl ester, CY5.5. A CY5.5 fluorescence labeling agent can be couple to the amine group of lysine linked to the PSMA ligand via a carbon spacer arm linker. A carbon spacer arm can include a standard (C6) or a longer spacer arm, such as a (Cl 2) spacer arm. In an exemplary embodiment, the carbon spacer arm is a C6 spacer arm

[0071] Chelating groups (with or without a chelated metal group) can include those disclosed in U.S. Patent No. 7,351,401, which is herein incorporated by reference in its entirety.

[0072] Near infrared imaging groups are disclosed in, for example, Tetrahedron Letters 49(2008) 3395-3399; Angew. Chem. Int. Ed. 2007, 46, 8998-9001; Anal. Chem. 2000, 72, 5907; Nature Biotechnology vol 23, 577-583; Eur Radiol(2003) 13: l95-208;and Cancer 67: 1991 2529-2537, which are herein incorporated by reference in their entirety.

[0073] Quantum dots, e.g., semiconductor particles, can be employed as described in Gao, et al "In vivo cancer targeting and imaging with semiconductor quantum dots", Nature Biotechnology, 22, (8), 2004, 969-976, the entire teachings of which are incorporated herein by reference. The PSMA ligands described herein can be coupled to the quantum dots, administered to a subject or a sample, and the subject/sample examined by spectroscopy or imaging to detect the labeled compound.

[0074] Magnetic resonance imaging (MRI) contrast agents can include positive contrast agents and negative contrast agents. The PSMA ligands described herein can be coupled to the MRI agents, administered to a subject or a sample, and the subject/sample examined by MRI or imaging to detect the labeled compound. Positive contrast agents (typically appearing predominantly bright on MRI) can include typically small molecular weight organic compounds that chelate or contain an active element having unpaired outer shell electron spins, e.g., gadolinium, manganese, iro, or the like. Typical contrast agents include gadopentetate dimeglumine, gadoteridol, gadoterate meglumine, mangafodipir trisodium, gadodiamide, and others known to the art. Negative contrast agents (typically appearing predominantly dark on MRI) can include small particulate aggregates comprised of superparamagnetic materials, for example, particles of superparamagnetic iron oxide (SPIO). Negative contrast agents can also include compounds that lack the hydrogen atoms associated with the signal in MRI imaging, for example, perfluorocarbons (perfluorochemicals).

[0075] In some embodiments, a PSMA targeted conjugate compound that is coupled to a fluorescence labeling agent or infrared agent can have the following general formula:

wherein Y is selected from at least one of a detectable moiety, therapeutic agent, or theranostic agent. This PSMA targeted conjugate compound can be prepared by linking the cysteine residue of the PSMA ligand to a lysine residue via a C6 linker, which then allows for coupling a second detectable moiety, therapeutic agent, or theranostic agent, for example, the fluorescence labeling agent, CY5.5 to the amine on the lysine amino acid.

[0076] In an exemplary embodiment, a PSMA targeted conjugate compound that is coupled to a fluorescence labeling agent and a therapeutic agent can have the formula:

[0077] In some aspects, the PSMA ligands coupled to a detectable moiety described herein may be used in conjunction with non-invasive imaging techniques for in vivo imaging, such as magnetic resonance spectroscopy (MRS) or imaging (MRI), or gamma imaging, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT), to determine the location or distribution of cancer cells. The term "in vivo imaging" refers to any method, which permits the detection of a labeled PSMA targeted conjugate compound, as described above. For gamma imaging, the radiation emitted from the organ or area being examined is measured and expressed either as total binding or as a ratio in which total binding in one tissue is normalized to (for example, divided by) the total binding in another tissue of the same subject during the same in vivo imaging procedure. Total binding in vivo is defined as the entire signal detected in a tissue by an in vivo imaging technique without the need for correction by a second injection of an identical quantity of molecular probe along with a large excess of unlabeled, but otherwise chemically identical compound.

[0078] For purposes of in vivo imaging, the type of detection instrument available is a major factor in selecting a given detectable moiety. For instance, the type of instrument used will guide the selection of the stable isotope. The half-life should be long enough so that it is still detectable at the time of maximum uptake by the target, but short enough so that the host does not sustain deleterious effects.

[0079] The PSMA targeted conjugate compounds coupled to a detectable moiety described herein can be administered to the subject by, for example, systemic, topical, and/or parenteral methods of administration· These methods include, e.g., injection, infusion, deposition, implantation, or topical administration, or any other method of administration where access to the tissue desired. In one example, administration can be by intravenous injection in the subject. Single or multiple administrations of the probe can be given.

“Administered”, as used herein, means provision or delivery is in an amount(s) and for a period of time(s) effective to label cancer cells in the subject.

[0080] The PSMA targeted conjugate compounds coupled to a detectable moiety described herein can be administered to a subject in a detectable quantity of a pharmaceutical composition containing the PSMA targeted conjugate compounds coupled to a detectable moiety described herein or a pharmaceutically acceptable water-soluble salt thereof, to a patient. A "detectable quantity" means that the amount of the detectable compound that is administered is sufficient to enable detection of binding of the compound to the cancer cells. An "imaging effective quantity" means that the amount of the detectable compound that is administered is sufficient to enable imaging of binding of the compound to the cancer cells.

[0081] The PSMA targeted conjugate compounds coupled to a detectable moiety described herein administered to a subject can be used to determine the presence, location, and/or distribution of cancer cells, i.e., PSMA expressing cancer cells or PSMA expressing neovaculature of the cancer cells, in an organ or body area of a patient. The presence, location, and/or distribution of the PSMA ligands coupled to a detectable moiety in the animal’s tissue, e.g., brain tissue, can be visualized (e.g., with an in vivo imaging modality described above). “Distribution” as used herein is the spatial property of being scattered about over an area or volume. In this case,“the distribution of cancer cells” is the spatial property of cancer cells being scattered about over an area or volume included in the animal’s tissue, e.g., prostate tissue. The distribution of the PSMA targeted conjugate compounds coupled to a detectable moiety may then be correlated with the presence or absence of cancer cells in the tissue. A distribution may be dispositive for the presence or absence of a cancer cells or may be combined with other factors and symptoms by one skilled in the art to positively detect the presence or absence of migrating or dispersing cancer cells, cancer metastases or define a tumor margin in the subject.

[0082] In some embodiments, the PSMA targeted conjugate compounds coupled to a detectable moiety may be administered to a subject to assess the distribution prostate cells in a subject and correlate the distribution to a specific location. Surgeons routinely use stereotactic techniques and intra-operative MRI (iMRI) in surgical resections. This allows them to specifically identify and sample tissue from distinct regions of the tumor such as the tumor edge or tumor center. Frequently, they also sample regions of prostate on the tumor margin that are outside the tumor edge that appear to be grossly normal but are infiltrated by dispersing tumor cells upon histological examination·

[0083] The PSMA targeted conjugate compounds coupled to a detectable moiety can be used in intra-operative imaging techniques to guide surgical resection and eliminate the “educated guess” of the location of the tumor by the surgeon. Previous studies have determined that more extensive surgical resection improves patient survival. Thus, targeted conjugate compounds coupled to a detectable moiety that function as diagnostic molecular imaging agents have the potential to increase patient survival rates.

[0084] The PSMA targeted conjugate compounds can be used to deliver a therapeutic anti-cancer agent to a PSMA expressing cancer cell of a subject or mammal. Therefore, some embodiments relate to the administration of a PSMA targeted conjugate compound coupled to an anticancer agent to a subject having or suspected of having cancer, such as a PSMA expressing cancer for the treatment of cancer. In particular embodiments, a PSMA targeted conjugate compounds coupled to an anticancer agent can be administered to a subject having prostate cancer.

[0085] The PSMA targeted conjugate compounds can target and transiently interact with, bind to, and/or couple with a cancer cell, such as a prostate cancer cell, and once interacting with, bound to, or coupled to the targeted cell or tissue advantageously facilitate delivery of a therapeutic agent within cell by, for example, receptor mediated endocytosis. Without being bound by theory, where the PSMA targeted conjugate compound includes a therapeutic agent, it is believed that once delivered to a PSMA expressing cell, endosomal conditions can release the therapeutic agent, e.g., doxorubicin, where it travels to the nucleus, binds to DNA and exerts anti-proliferative effects.

[0086] Examples of anticancer agents that can be directly or indirectly coupled to a PSMA targeting conjugate compound as described herein can include, but are not limited to Taxol, Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin;

ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium;

bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflomithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; fluorocitabine; fosquidone; fostriecin sodium; gemcitabine;

gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or rIL2), interferon a-2a; interferon a-2b; interferon a-nl; interferon a-n3; interferon b-I a; interferon g-I b; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine;

mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper;

mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin;

ormaplatin; oxisuran; pegaspargase; peliomycin; pentamustine; peplomycin sulfate;

perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safingol

hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.

[0087] Other exemplary anti-cancer agents include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox;

amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole;

andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti- dorsalizing morphogenetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen;

antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists;

benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine;

calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine;

carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4;

combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8;

cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone;

didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; 9-dioxamycin; diphenyl spiromustine; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur;

epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists;

etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunomnicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin- like growth factor- 1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B ; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon;

leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine;

lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A;

marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor;

mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor l-based therapy; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N- substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; 06-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; palauamine;

palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B;

plasminogen activator inhibitor; platinum complex; platinum compounds; platinum- triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone Bl; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A;

sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense

oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen-binding protein; silicon phthalocyanine (PC4) sizofuran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin;

sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine;

synthetic glycosaminoglycans (GAGs); tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist;

thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.

[0088] Other exemplary anti-cancer agents can include the following marketed drugs and drugs in development: Erbulozole (also known as R-55104), Dolastatin 10 (also known as DLS-10 and NSC-376128), Mivobulin isethionate (also known as CI-980), Vincristine, NSC-639829, Discodermolide (also known as NVP-XX-A-296), ABT-751 (Abbott, also known as E-7010), Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C), Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9), Cemadotin hydrochloride (also known as LU- 103793 and NSC-D-669356), Epothilones (such as Epothilone A, Epothilone B, Epothilone C (also known as desoxyepothilone A or dEpoA), Epothilone D (also referred to as KOS-862, dEpoB, and desoxyepothilone B), Epothilone E, Epothilone F, Epothilone B N-oxide, Epothilone A N-oxide, l6-aza-epothilone B, 2l-aminoepothilone B (also known as BMS-310705), 21 -hydroxy epothilone D (also known as Desoxyepothilone F and dEpoF), 26-fluoroepothilone), Auristatin PE (also known as NSC-654663), Soblidotin (also known as TZT-1027), LS-4559-P (Pharmacia, also known as LS-4577), LS-4578 (Pharmacia, also known as LS-477-P), LS-4477 (Pharmacia), LS-4559 (Pharmacia), RPR- 112378 (Aventis), Vincristine sulfate, DZ-3358 (Daiichi), FR-182877 (Fujisawa, also known as WS-9885B), GS-164 (Takeda), GS-198 (Takeda), KAR-2 (Hungarian Academy of Sciences), BSF-223651 (BASF, also known as ILX-651 and LU-223651), SAH-49960 (Lilly/Novartis), SDZ-268970 (Lilly/Novartis), AM-97 (Armad/Kyowa Hakko), AM- 132 (Arnad), AM-138 (Armad/Kyowa Hakko), IDN-5005 (Indena), Cryptophycin 52 (also known as LY-355703), AC-7739 (Ajinomoto, also known as AVE-8063A and CS-39.HC1), AC- 7700 (Ajinomoto, also known as AVE-8062, AVE-8062A, CS-39-L-Ser.HCl, and RPR- 258062A), Vitilevuamide, Tubulysin A, Canadensol, Centaureidin (also known as NSC- 106969), T-138067 (Tularik, also known as T-67, TL-138067 and TI-138067), COBRA-l (Parker Hughes Institute, also known as DDE-261 and WHI-261), H10 (Kansas State University), H16 (Kansas State University), Oncocidin Al (also known as BTO-956 and DIME), DDE-313 (Parker Hughes Institute), Fijianolide B, Laulimalide, SPA-2 (Parker Hughes Institute), SPA-l (Parker Hughes Institute, also known as SPIKET-P), 3-IAABU (Cytoskeleton/Mt. Sinai School of Medicine, also known as MF-569), Narcosine (also known as NSC-5366), Nascapine, D-24851 (Asta Medica), A-105972 (Abbott), Hemiasterlin, 3- BAABU (Cytoskeleton/Mt. Sinai School of Medicine, also known as MF-191), TMPN (Arizona State University), Vanadocene acetylacetonate, T-138026 (Tularik), Monsatrol, Inanocine (also known as NSC-698666), 3-IAABE (Cytoskeleton/Mt. Sinai School of Medicine), A-204197 (Abbott), T-607 (Tularik, also known as T-900607), RPR-l 15781 (Aventis), Eleutherobins (such as Desmethyleleutherobin, Desaetyleleutherobin,

Isoeleutherobin A, and Z-Eleutherobin), Caribaeoside, Caribaeolin, Halichondrin B, D-64131 (Asta Medica), D-68144 (Asta Medica), Diazonamide A, A-293620 (Abbott), NPI-2350 (Nereus), Taccalonolide A, TUB-245 (Aventis), A-259754 (Abbott), Diozostatin, (-)- Phenylahistin (also known as NSCL-96F037), D-68838 (Asta Medica), D-68836 (Asta Medica), Myoseverin B, D-43411 (Zentaris, also known as D-81862), A-289099 (Abbott), A- 318315 (Abbott), HTI-286 (also known as SPA-110, trifluoroacetate salt) (Wyeth), D-82317 (Zentaris), D-82318 (Zentaris), SC-12983 (NC1), Resverastatin phosphate sodium, BPR-OY- 007 (National Health Research Institutes), and SSR-250411 (Sanofi).

[0089] Still other exemplary anti-cancer agents include alkylating agents, such as nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc.), ethylenimine and methylmelamines (e.g., hexamethlymelamine, thiotepa), alkyl sulfonates (e.g., busulfan), nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc.), or triazenes (decarbazine, etc.), antimetabolites, such as folic acid analog

(e.g., methotrexate), or pyrimidine analogs (e.g., fluorouracil, floxouridine, Cytarabine), purine analogs (e.g., mercaptopurine, thioguanine, pentostatin, vinca alkaloids

(e.g., vinblastin, vincristine), epipodophyllotoxins (e.g., etoposide, teniposide), platinum coordination complexes (e.g., cisplatin, carboblatin), anthracenedione (e.g., mitoxantrone), substituted urea (e.g., hydroxyurea), methyl hydrazine derivative (e.g., procarbazine), adrenocortical suppressant (e.g., mitotane, amino glutethimide).

[0090] In certain embodiments, the anti-cancer agent coupled at Y or Y 1 to a PSMA targeted conjugate compound is selected from doxorubicin or monomethyl auristatin E (MMAE). In particular embodiments, the anti-cancer agent is coupled at Y to the PSMA targeted conjugate compound. In an exemplary embodiment, a PSMA targeted conjugate compound that is coupled to a therapeutic anti-cancer agent can have the formula:

r pharmaceutical salts thereof. [0091] The PSMA targeting conjugate compounds can be administered alone as a monotherapy, or in conjunction with or in combination with one or more additional therapeutic agents. In an exemplary embodiment, a PSMA targeting conjugate compound described herein can be administered to the subject in combination with an additional anti cancer agent. In a particular embodiment, a PSMA targeting conjugate compound is coupled to doxorubicin as described herein can be administered to the subject in combination with the additional anti-cancer agent docetaxel.

[0092] It will be appreciated that additional detectable moieties, therapeutic agents, and/or theranostic agents administered to a subject need not be conjugated directly or indirectly to the PSMA ligand of the PSMA targeting compound and can optionally be provided in a pharmaceutical composition or preparation with the PSMA targeting conjugate compounds described herein or in a separate pharmaceutical composition.

[0093] The term "in conjunction with" or“in combination with” indicates that the PSMA targeting conjugate compound is administered at about the same time as the additional agent. The PSMA targeting conjugate compound can be administered to the subject in need thereof as part of a pharmaceutical composition comprising the compound and a

pharmaceutically acceptable carrier or excipient and, optionally, one or more additional therapeutic agents. The compound and additional therapeutic agent can be components of separate pharmaceutical compositions, which can be mixed together prior to administration or administered separately. The PSMA targeting conjugate compound can, for example, be administered in a composition containing the additional therapeutic agent, and thereby, administered contemporaneously with the agent. Alternatively, the PSMA targeting conjugate compound can be administered contemporaneously, without mixing (e.g., by delivery of the compound on the intravenous line by which the compound is also

administered, or vice versa). In another embodiment, the PSMA targeting conjugate compound can be administered separately (e.g., not admixed), but within a short timeframe (e.g., within 24 hours) of administration of the compound.

[0094] The disclosed PSMA targeting conjugate compounds and additional therapeutic agents, detectable moieties, and/or theranostic agents described herein can be administered to a subject by any conventional method of drug administration, for example, orally in capsules, suspensions or tablets or by parenteral administration. Parenteral administration can include, for example, intramuscular, intravenous, intraventricular, intraarterial, intrathecal, subcutaneous, or intraperitoneal administration. The disclosed compounds can also be administered orally (e.g., in capsules, suspensions, tablets or dietary), nasally (e.g., solution, suspension), transdermally, intradermally, topically (e.g., cream, ointment), inhalation (e.g., intrabronchial, intranasal, oral inhalation or intranasal drops) transmucosally or rectally. Delivery can also be by injection into the brain or body cavity of a patient or by use of a timed release or sustained release matrix delivery systems, or by onsite delivery using micelles, gels and liposomes. Nebulizing devices, powder inhalers, and aerosolized solutions may also be used to administer such preparations to the respiratory tract. Delivery can be in vivo, or ex vivo. Administration can be local or systemic as indicated. More than one route can be used concurrently, if desired. The preferred mode of administration can vary depending upon the particular disclosed compound chosen. In specific embodiments, oral, parenteral, or systemic administration (e.g., intravenous) are preferred modes of

administration for treatment.

[0095] The methods described herein contemplate either single or multiple

administrations, given either simultaneously or over an extended period of time. The PSMA targeting conjugate compound (or composition containing the compound) can be

administered at regular intervals, depending on the nature and extent of the inflammatory disorder's effects, and on an ongoing basis. Administration at a "regular interval," as used herein, indicates that the therapeutically effective amount is administered periodically (as distinguished from a one-time dose). In one embodiment, the PSMA targeting conjugate compound is administered periodically, e.g., at a regular interval (e.g., bimonthly, monthly, biweekly, weekly, twice weekly, daily, twice a day or three times or more often a day).

[0096] The administration interval for a single individual can be fixed, or can be varied over time, depending on the needs of the individual. For example, in times of physical illness or stress, or if disease symptoms worsen, the interval between doses can be decreased.

Depending upon the half-life of the agent in the subject, the agent can be administered between, for example, once a day or once a week.

[0097] For example, the administration of the PSMA targeting conjugate compound described herein and/or the additional therapeutic agent can take place at least once on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28,

29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least once on week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, or any combination thereof, using single or divided doses of every 60, 48, 36, 24, 12, 8, 6, 4, or 2 hours, or any combination thereof. Administration can take place at any time of day, for example, in the morning, the afternoon or evening. For instance, the administration can take place in the morning, e.g., between 6:00 a.m. and 12:00 noon; in the afternoon, e.g., after noon and before 6:00 p.m.; or in the evening, e.g., between 6:01 p.m. and midnight.

[0098] A disclosed PSMA targeting conjugate compound and/or additional therapeutic agent can be administered in a dosage of, for example, 0.1 to 100 mg/kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26,

27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day. Dosage forms (composition) suitable for internal administration generally contain from about 0.1 milligram to about 500 milligrams of active ingredient per unit. In these pharmaceutical compositions, the active ingredient will ordinarily be present in an amount of about 0.5-95% by weight based on the total weight of the composition.

[0099] The amount of disclosed PSMA targeting conjugate compound and/or additional therapeutic agent administered to the subject can depend on the characteristics of the subject, such as general health, age, sex, body weight and tolerance to drugs as well as the degree, severity and type of rejection. The skilled artisan will be able to determine appropriate dosages depending on these and other factors using standard clinical techniques.

[00100] In addition, in vitro or in vivo assays can be employed to identify desired dosage ranges. The dose to be employed can also depend on the route of administration, the seriousness of the disease, and the subject's circumstances. Effective doses may be extrapolated from dose -response curves derived from in vitro or animal model test systems. The amount of the compound can also depend on the disease state or condition being treated along with the clinical factors and the route of administration of the compound.

[00101] For treating humans or animals, the amount of disclosed PSMA targeting conjugate compound and/or additional therapeutic agent administered (in milligrams of compound per kilograms of subject body weight) is generally from about 0.1 mg/kg to about 100 mg/kg, typically from about 1 mg/kg to about 50 mg/kg, or more typically from about 1 mg/kg to about 25 mg/kg. In a preferred embodiment, the effective amount of agent or PSMA targeting conjugate compound is about 1-10 mg/kg. In another preferred

embodiment, the effective amount of agent or PSMA targeting conjugate compound is about 1-5 mg/kg. The effective amount for a subject can be varied (e.g., increased or decreased) over time, depending on the needs of the subject.

[00102] The term "unit dose" refers to a physically discrete unit suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material that can produce the desired therapeutic effect in association with the required diluent; e.g., carrier or vehicle. In addition to the ingredients particularly mentioned above, the formulations of the present invention may include other agents conventional in the art having regard to the type of formulation in question.

[00103] The disclosed PSMA targeting conjugate compound and/or additional therapeutic agent described herein can be administered to the subject in conjunction with an acceptable pharmaceutical carrier or diluent as part of a pharmaceutical composition for therapy. Formulation of the PSMA targeting conjugate compound to be administered will vary according to the route of administration selected (e.g., solution, emulsion, capsule, and the like). Suitable pharmaceutically acceptable carriers may contain inert ingredients that do not unduly inhibit the biological activity of the PSMA targeting conjugate compounds. The pharmaceutically acceptable carriers should be biocompatible, e.g., non-toxic, non inflammatory, non-immunogenic and devoid of other undesired reactions upon the administration to a subject. Standard pharmaceutical formulation techniques can be employed, such as those described in Remington's Pharmaceutical Sciences, ibid. Suitable pharmaceutical carriers for parenteral administration include, for example, sterile water, physiological saline, bacteriostatic saline (saline containing about 0.9% mg/ml benzyl alcohol), phosphate-buffered saline, Hank's solution, Ringer's-lactate and the like. Methods for encapsulating compositions (such as in a coating of hard gelatin or cyclodextran) are known in the art (Baker, et a , "Controlled Release of Biological Active Agents", John Wiley and Sons, 1986).

[00104] The preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art. Typically, such compositions are prepared as injectables as either liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared. Formulation will vary according to the route of administration selected (e.g., solution, emulsion, capsule).

[00105] A pharmaceutically acceptable carrier for a pharmaceutical composition can also include delivery systems known to the art for entraining or encapsulating drugs, such as anticancer drugs. In some embodiments, the disclosed compounds can be employed with such delivery systems including, for example, liposomes, nanoparticles, nanospheres, nanodiscs, dendrimers, and the like. See, for example Farokhzad, O. C., Jon, S.,

Khademhosseini, A., Tran, T. N., Lavan, D. A., and Langer, R. (2004). "Nanoparticle- aptamer bioconjugates: a new approach for targeting prostate cancer cells." Cancer Res., 64, 7668-72; Dass, C. R. (2002). "Vehicles for oligonucleotide delivery to tumours." J. Pharm. Pharmacol., 54, 3-27; Lysik, M. A., and Wu-Pong, S. (2003). "Innovations in oligonucleotide drug delivery." J. Pharm. Sci., 92, 1559-73; Shoji, Y., and Nakashima, H. (2004). "Current status of delivery systems to improve target efficacy of oligonucleotides." Curr. Pharm. Des., 10, 785-96; Allen, T. M., and Cullis, P. R. (2004). "Drug delivery systems: entering the mainstream." Science, 303, 1818-22. The entire teachings of each reference cited in this paragraph are incorporated herein by reference.

[00106] The following examples are for the purpose of illustration only and are not intended to limit the scope of the claims, which are appended hereto.

Example 1

PSMA-l Doxorubicin Conjugates for Targeted Therapy of Prostate Cancer

[00107] We have developed a highly negatively-charged peptide-based PSMA receptor ligand, PSMA-l, with affinity in the nM range. Here we aim to capitalize on the targeting ability of our PSMA-l ligand, in order to improve the efficacy and widen the therapeutic window of the commonly used chemotherapeutic agent, doxorubicin.

[00108] Clinically, doxorubicin’ s use is not optimal due to significant toxicities including cardiotoxicity and myelosuppression, effects which may be decreased with improved tumor-targeting.

[00109] We attached PSMA-l to doxorubicin with different linkages that can be cleaved via proteases or the acidic environment found at/within most tumors. With this prodrug strategy, demonstrated in Fig. 1 and Fig. 2, doxorubicin can dissociate from PSMA-l inside of a cancer cell or in the extracellular milieu and exert its anti-proliferative effects freely. Further, we will exploit the inactivity of conjugated doxorubicin to decrease off target toxicity typically found with the free or liposome-incorporated drug. In summary, we show PSMA-l -Doxorubicin conjugates will be selectively taken up by PSMA-expressing prostate cancer cells, and these drugs exhibit equal or superior antineoplastic effects in vitro and in vivo compared to free doxorubicin with minimal off-target toxicity.

[00110] Fig. 1 illustrates our reaction schema, in brief. We use a series of three linkers to join the PSMA-l ligand to doxorubicin:

1. Non-cleavable linker, SMCC (succinimidyl 4-(N- maleimidomethyl)cyclohexane-l-carboxylate). Use of this linker does not yield a true prodrug as with the other two linkers below.

2. Protease-cleavable linker, MC-Val-Cit-PABC-PNP

(Maleimidocaproyl-L-Valine-L-Citrulline-p-Aminobenzyl Alcohol p-Nitrophenyl

Carbonate). This is the linker utilized in the FDA approved antibody-drug conjugate hrentuximah vedotin for treatment of Hodgkin’ s disease, and consists of MC and PABC spacers sandwiching the cathepsin B-sensitive dipeptide, valine-citrulline. Cathepsin B cleaves the valine-citrulline bond, the PABC spacer is self-imolative, and intact doxorubicin in its native state should be released.

3. Acid-labile hydrazone linker, MMCCH (4-(4- maleimidomethyljcyclohexane- 1 -carboxyl hydrazide) (43). The hydrozone bond cleaves releasing doxorubicin in its native state.

Methods

Materials

[00111] Fmoc rink amide MB HA resin and Fmoc amino acids were purchased from Peptide International Inc, Novabiochem, and Advanced Biochemical Compounds.

Doxorubicin HC1 was purchased from LC Laboratories. SMCC was purchased from ThermoFisher. MC-Val-Cit-PABC-PNP was purchased from MedChem Express. MMCCH TFA was purchased from Chem-Impex International Inc. Basic solvents and laboratory supplies were purchased from ThermoFisher and Sigma Aldrich.

General Comments

[00112] Preparative high-performance liquid chromatography (HPLC) was used for all purifications, unless otherwise noted. A Shimadzu HPLC system was utilized with a UV/visible detector monitored at 220 nm. A Luna 5m Cis(2) 100A column

(250mmxl0mmx5pm, Phenomenex, Torrance, CA) was used at a flow rate of 2.5ml/min. Gradients and buffers as noted below. Svnthesis of PSMA-l (Cvs) (Glu-CO-Glu’-Amc-Ahx-Glu-Glu-Glu-Cvs-NH2)

[00113] PSMA-l(Cys) was synthesized as PSMA-l has been described before, with a substitution of cysteine in place of the terminal lysine to provide the -SH group necessary for reaction with the maleimide group of each linker. PSMA-l (Cys) was synthesized manually using standard Fmoc chemistry. Generally, peptide was synthesized at 0.2 mmol scale starting from C-terminal Fmoc-rink amide MB HA resin.

[00114] Resin was allowed to soak in dimethylformamide (DMF) overnight before beginning synthesis. Fmoc deprotection initially and for each coupling cycle was carried out using 20% piperidine in DMF, applied to resin and shaken for 5 minutes. Coupling reactions were carried out using 3.3 equivalents of Fmoc amino acids in DMF activated with 3.3 equivalents of 2-(6-chloro- lH-benzotriazole- 1 -yl)- 1 , 1 ,3,3-tetramethylaminium

hexafluorophosphate (HCTU) and 5 equivalents of diisopropylethylamine (DIPEA) in DMF, with this mixture applied to the resin and shaken for 30 minutes. These alternating steps (deprotection, new peptide, repeat) were used to build the peptide sequence Fmoc-Glu'-Amc- Ahx-Glu-Glu-Glu-Cys(Mtt) on the resin.

[00115] The Fmoc group of N-terminal amino acid Glu' was deprotected by 20% piperidine. A chloroform solution containing 3 equivalents of H-Glu(OtBu)-OtBu mixed with 2.5 equivalents of DIPEA was prepared. The solution was then added slowly to 0.25 equivalents triphosgene in chloroform over 10 minutes at room temperature. After 15 -minute incubation, the reaction blend was mixed with Glu'-Amc-Ahx-Glu-Glu-Glu-Lys on rink amide resin preswollen in chloroform with 2.5 equivalents of DIPEA. After the reaction was complete, the resin was washed with DMF and then dichloromethane and dried. The peptide was cleaved from resin by TFA/water/triisopropylsilane (950:25:25).

[00116] The cleaved peptide was purified by preparative high-performance liquid chromatography (HPLC). The gradient was 5-55% acetonitrile against 0.1% trifluoroacetic acid over 30 minutes, followed by 15 minutes of 55% acetonitrile. Peptide retention time was 21 minutes. Expected structures were confirmed by molecular weight using electrospray mass spectrometry (MS). Peptide was lyophilized and stored at -20°C.

Synthesis of PSMA-l -Doxorubicin Conjugates

[00117] Generally, each linker was reacted with an excess of doxorubicin in DMF and purified by HPLC, resulting in a maleimide-functionalized doxorubicin. Next, the cysteine tail of the PSMA ligand was reacted with the maleimide group on the linker to form the final conjugate. (Doxorubicin is light-sensitive and was handled with special care to wrap tubes in aluminum foil to prevent light exposure). The final conjugate was again purified by HPLC and lyophilized for storage at -20°C. The structures at each reaction step were confirmed using either electrospray MS or MALDI-MS. Our electrospray MS machine generally produced cleaner images and was used when available. MALDI was used in other cases or if MW of the compound exceeded 2200, the maximum electrospray capacity. Our PSMA- targeted conjugates do make use of the modified PSMA-l(Cys), but for simplicity they will be referred to as PSMA- 1 -Doxorubicin conjugates.

Synthesis of PSMA-l-SMCC-Dox

[00118] To produce the maleimide-functionalized doxorubicin, SMCC was dissolved in DMF with the addition of triethylamine (TEA) to adjust the solution pH to approximately 8, measured roughly with pH paper. To this, 1.1 equivalents of doxorubicin HC1 was added. The mixture was allowed to mix in the dark overnight at room temperature before purification by HPLC using a gradient of 30-90% acetonitrile against 0.1% trifluoroacetic acid over 40 minutes. Retention time for unreacted doxorubicin was 13 minutes, unreacted SMCC was 23.5 minutes, and the product SMCC-Dox was 24.5 minutes.

[00119] To produce PSMA-l-SMCC-Dox, SMCC-Dox was reacted with 1.1 equivalents PSMA-l(Cys) in DMF with pH adjustment to 8 by TEA. The solution was allowed to mix overnight in dark conditions at room temperature. The same HPLC gradient was used resulting in a retention time for the final product of 17 minutes. Unreacted PSMA-l(Cys) elutes with the solvent DMF. The reaction generally went to completion with no unreacted SMCC-Dox.

Synthesis of PSMA-l-MC-Val-Cit-PABC-Dox

[00120] To produce the maleimide-functionalized doxorubicin, MC-Val-Cit-PABC-PNP was reacted with 1.1 equivalents doxorubicin HC1 and 1.1 equivalents DIPEA in DMF. It was left to react on a spinner in the dark for two days at room temperature. This single reaction was purified without using HPLC. Instead, ~10c volume excess CH2C12 was added to the reaction mixture and this was left to sit overnight at -20 to precipitate the product MC- Val-Cit-PABC-Dox. The resultant solid was isolated by centrifugation. [00121] To produce PSMA-l-MC-Val-Cit-PABC-Dox, the MC-Val-Cit-PABC-Dox was reacted with 1.1 equivalents of PSMA-l(Cys) in DMF with pH adjusted to 7-8 using TEA. This was allowed to spin in the dark overnight. The product was purified with HPLC using a gradient of 30-90% acetonitrile against 0.1% trifluoroacetic acid over 40 minutes. Retention time was 17 minutes. Unreacted PSMA-l(Cys) elutes with the solvent DMF. This reaction generally went to completion with no unreacted MC-Val-Cit-PABC-Dox.

Synthesis of PSMA-l-MMCCH-Dox

[00122] To produce the maleimide-functionalized doxorubicin, MMCCH TFA was reacted with 1.1 equivalents doxorubicin HC1 in DMF at 50C for 2 hours in a dark fume hood. The product was purified with HPLC using a gradient of 30-90% acetonitrile against 2 mM TEAA. A basic buffer was chosen to avoid lysis of the acid-labile linkage. Retention time of the uncreated MMCCH was 8 minutes, unreacted doxorubicin was 15 minutes, and MMCC-Dox was 19 minutes.

[00123] To produce PSMA-l-MMCCH-Dox, MMCCH-Dox was reacted with 1.1 equivalents PSMA-l(Cys) in DMF with pH adjusted to 8 using TEA. This was mixed overnight and the product purified using an HPLC gradient of 10-90% acetonitrile against 2 mM TEAA. Product retention time was 19 minutes. Unreacted PSMA-l(Cys) elutes with the solvent DMF. This reaction generally went to completion with no unreacted MMCCH- Dox.

Cell Culture

[00124] Retrovirally transfected PSMA-positive PC3pip cells and control PC3flu cells were used for all experiments. Cells were cultured at 37C in a 5% CO2, humidified atmosphere. Cultures were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS).

In vitro Cellular Uptake Studies

[00125] PC3Pip and PC3flu cells (1.5 x 10 5 ) were plated on coverslips and allowed to adhere for 36-48 hours. Cells were incubated in 1 mM of free doxorubicin or the various PSMA-l -Doxorubicin conjugates in RPMI media. Similar experiments are reported widely in the literature, with doses usually in the pM range because of doxorubicin’ s relatively dim fluorescence, at least compared with some of the commercially available dyes used in our lab (44) (45). This dose was chosen to balance that relatively dim fluorescence with a goal of attempting to maintain selectivity.

[00126] The PSMA-l -Doxorubicin conjugates were not soluble in water. Stock solutions were prepared in dimethyl sulfoxide, DMSO, for short term storage. Experimental dilutions were prepared in RPMI media. An extinction coefficient of 10410 liter/(mol cm) was used, as reported in the literature, for dilution calculations using the Beer-Lambert Law after absorption was measured at 480 nm.

[00127] Next, cells were washed 3 times with RPMI media, fixed with 4%

paraformaldehyde for 10 minutes, and washed twice with phosphate-buffered saline (PBS). The cells were counterstained with DAPI nuclear staining, mounted on microscope slides with a 50% mounting solution mixture of PBS and glycol, and the coverslips were sealed with clear nail polish. Fluorescent imaging was performed with Leica DM4000B

fluorescence microscopy. DAPI imaging used the Blue filter with excitation/emission at 410/455 nm. Doxorubicin was imaged with the Texas Red filter with Ex/Em 587/612 nm.

Cytotoxicity Assay

[00128] PC3Pip and PC3Flu cells (2000 cells/lOO pL/well) were plated in 96 well plates and rested for 24 hours to adhere to the plate surface. To each well, we added 10 pL of an 11X concentrated solution of doxorubicin or conjugate. Each drug dose was tested in groups of six wells. Dojindo CCK8 assays were used to determine relative cell survival of treated wells compared with untreated controls. This is a calorimetric assay for determination of cell viability and proliferation. A water-soluble tetrazolium salt, WST-8, is reduced by dehydrogenase activity in the cell which produces an orange dye with relative amounts quantified by absorbance. Briefly, 10 pL assay solution was added to each well, the plate was left to incubate while orange color was produced, the absorption of the solution in the plate was read at 450 nm, and the cell viabilities were calculated.

Mouse Tumor Xenograft Studies

[00129] All animal procedures were performed according to the protocols approved by the Institutional Animal Care and Use Committee (IACUC). Our approval is Case Western Reserve University IACUC #2015-0033. Athymic nude mice aged 6-8 weeks were implanted subcutaneously with 1x106 PC3Pip cells in 100 pL Matrigel on the right flank. Tumors were allowed to grow for two weeks. Mice were then treated with three doses of free doxorubicin or PSMA-l-MMCCH-Dox in PBS on day 0, 7, and 14 of the study via tail vein injection. Dose used was 2 mg/kg doxorubicin, or the equivalent molar dosage of the PSMA- targeted prodrug. Tumor dimensions were measured using calipers twice per week, and mouse weights were recorded on these days, as well. The study duration was 21 days.

Statistical Analysis

[00130] Statistical analysis was performed using student t-test with a p<0.05 meeting criteria for statistical significance. Analysis was performed using JMP Pro 13 software.

Results

Chemistry

[00131] PSMA-l(Cys) is a modified version of a PSMA-l peptide ligand. It is a urea- based ligand with three glutamic acid residues providing a negative charge that has been previously shown to reduce non-specific background binding without negatively influencing PSMA affinity. The terminal cysteine allows for easy coupling of the -SH group to the maleimide in each of the linkers chosen. PSMA-l(Cys) was successfully synthesized by Fmoc chemistry by hand, with an electrospray MS peak showing 1062.4 g/mol. This was consisted with the calculated molecular weight using Chem Draw Professional of

1062.1 g/mol, as demonstrated in Fig. 3.

[00132] The non-cleavable PSMA-l-SMCC-Dox conjugate was synthesized by allowing the NHS ester in SMCC to react with the -NH2 present in doxorubicin. Expected molecular weight for this compound was 762.3. MALDI-MS of SMCC-Dox showed a strong peak at 785.4, consistent with MW+Na, and confirming successful conjugation. The full PSMA-l- SMCC-Dox conjugate was also successfully produced when the -SH of cysteine in the linker combined with the maleimide end of the SMCC linker. The expected molecular weight for this compound was 1824.9. See Figs. 4 and 5.

[00133] Similarly, the production of the protease-cleavable PSMA-l-MC-Val-Cit- PABC-Dox was achieved by reaction of the NHS ester group in the MC-Val-Cit-PABC-PNP linker with the amine in doxorubicin. The expected MW was 1142.2, and MALDI-MS demonstrated a peak at 1164.2, consistent with MW+Na. The -SH in PSMA-l(Cys) reacts nicely with the maleimide group of the linker to produce the final product, with MALDI-MS showing a peak at 2225.6, again consistent with MW+NA (expected MW 2204.3). See Figs.

6 and 7 for reaction data.

[00134] The final conjugate, acid-labile PSMA-l-MMCCH-Dox, was produced by first generating MMCCH-Dox. This is another maleimide-functionalized doxorubicin, but with a hydrazone bond at the carbonyl group of doxorubicin rather than an NHS ester to amine reaction. Electrospray MS shows a peak at the expected molecular weight of 777 and a strong peak at 799, consistent with MW+Na. There are also peaks at 1553.3 consistent with a dimer and 1575.4 consistent with the dimer+Na. As above, the -SH in the PSMA ligand joins the maleimide in the MMCCH linker to produce the final product. See Figs. 8 and 9.

[00135] Conjugation of the PSMA-targeting ligand to doxorubicin via the three linkers tested did not influence the absorption spectra of doxorubicin. Peak absorption was seen at 480 nm for free doxorubicin and each conjugate, as demonstrated in Fig. 10.

In vitro Cellular Uptake Studies

[00136] To determine whether the PSMA- 1 -Doxorubicin conjugates might be preferentially uptaken by PSMA-expressing PC3Pip cells compared to PSMA-negative PC3Flu cells, both of these cell lines were treated with 1 mM of doxorubicin or PSMA-l- Doxorubicin conjugate for either 90 minutes or 48 hours. Doxorubicin fluoresces and can therefore be visualized in cells by fluorescent microscopy. Qualitative results are shown in Fig. 11. Free doxorubicin was equally seen in PC3Pip and PC3Flu cell lines, and the drug was seen primarily in the nucleus, overlying the DAPI nuclear staining in these cells. PSMA- l-SMCC-Dox demonstrated more signal in PC3Pip compared to PC3Flu cells, and as was the case with all PSMA-conjugates, the selectivity was more obvious at short cell treatment times. However, unlike free doxorubicin, the signal was seen in a spotty, endosomal type pattern. The same observations were true of PSMA-l-Val-Cit-PABC-Dox. PSMA-l- MMCCH-Dox showed similar selectivity for PC3Pip cells, but the doxorubicin signal did colocalize with the DAPI nuclear staining for this conjugate.

In vitro Cytotoxicity

[00137] To determine whether our PSMA- 1 -Doxorubicin conjugates demonstrated any cytotoxic effect in vitro, and whether they may be more toxic to PSMA-expressing cells, the commercially available Dojindo CCK8 assay was used to monitor cell viability after treatment with free and PSMA-targeted doxorubicin. First, PC3Pip and PC3Flu cells were treated continuously for 48 hours with free doxorubicin, non-cleavable PSMA-l-SMCC-Dox, cathepsin-cleavable PSMA-l-MC-Val-Cit-PABC-Dox, and acid-labile PSMA-l-MMCCH- Dox. Doxorubicin exhibited potent, equal growth inhibition of PC3Pip and PC3Flu cell lines. PSMA-l-SMCC-Dox and PSMA-l-MC-Val-Cit-PABC-Dox showed essentially no growth inhibition of either cell line. PSMA-l-MMCCH-Dox, however, was significantly more toxic to PC3Pip cells compared to PC3Flu cells at all doses tested over the range 62.5- 2000 nM, as depicted in Fig. 12.

[00138] The fluorescence microscopy data indicated that some PSMA-l-MMCCH-Dox does enter PC3Flu cells after long incubation periods, so the cytotoxicity assay was also repeated for free doxorubicin and the acid-labile conjugate at a slightly shorter incubation period of 24 hours in an attempt to optimize the cytotoxicity difference in the two cell lines. The cells were then provided fresh media and allowed to continue in culture for an additional 48 hours to allow for sufficient time for growth inhibition and/or cell death. The increased toxicity to PC3Pip cells compared to PC3Flu cells was again evident, at about the same magnitude, for these conditions. The drug conjugate, in vitro, was generally less toxic than free drug, especially at lower doses, as seen in Fig. 13B below which shows ratios of PSMA- l-MMCCH-Dox toxicity to free doxorubicin viability. At doses of 500 nM or more, the PSMA-l-MMCCH-Dox conjugate showed equal or superior cytotoxicity compared to free drug for PC3Pip cells, and at 1000 nM or more the targeted drug was also toxic to PC3Flu cells similarly to free doxorubicin.

Mouse Tumor Xenograft Response

[00139] Mice implanted with PSMA(+) PC3Pip flank tumors were treated with three weekly doses of 2 mg/kg of free doxorubicin, or the molar equivalent of PSMA-l-MMCC- Dox, 6.35 mg/kg. Tumor size was monitored with calipers, and toxicity was measured by mouse weight over time. Measurements were taken two times per week, day 0, 4, 7, 11, etc. until day 21. At these tested doses, all tumors grew in size with time. The mice treated with free doxorubicin exhibited more rapid tumor growth than those treated with PSMA-l- MMCCH-Dox, with growth difference reaching statistical significance through day 14 and trending toward significance through the end of the study. Additionally, mice treated with the targeted drug exhibited less weight loss, an indication of drug toxicity, over the duration of the study (1.8% vs. 17.3%, p=0.009), as depicted in Fig. 14 below.

[00140] At this time, PSMA-l-SMCC-Dox and PSMA-l-MC-Val-Cit-PABC-Dox have not been tested in mice. We would hypothesize that the conjugate with non-cleavable SMCC linker would be targeted but not very effective, as it was in vitro. The cathepsin-cleavable drug, though, may show better effect in vivo if cathepsin expression is higher in the tumor model than in cell culture. Further, though there is sufficient evidence to conclude that PSMA-l-MMCCH-Dox is more effective than free doxorubicin, dosing studies should be performed to determine maximum-tolerated dose and to see if tumor shrinkage can be achieved.

[00141] In conclusion, three PSMA-l -Doxorubicin conjugates were synthesized making use of differing linker chemistries. All three were selective for PSMA(+) cell lines, but only acid-labile PSMA-l-MMCCH-Dox promoted release of free doxorubicin, allowing the drug to travel to the nucleus and exert its cytotoxic activity. This drug was tested in a flank tumor prostate cancer model, and resulted in tumor growth inhibition compared to free doxorubicin. Further, it was less toxic to the mice than free doxorubicin as determined by decreased weight loss while on treatment. It is anticipated that these agents may prove more effective while reducing significant toxicities/side effects compared with free doxorubicin for treatment of metastatic prostate cancer models in nude mice and later for treatment of human patients with prostate cancer or other PSMA-expressing tumors.

Example 2

PSMA-MMAE-Cv5.5 Conjugates for Targeted Therapy of Prostate Cancer

[00142] Monomethylauristatin E (MMAE) is a synthetic analogue of dolastatin-lO, a potent inhibitor of microtubule polymerization that was originally isolated from the Indian Ocean sea hare Dolabella auricularia. Problems with toxicity have limited its effectiveness as an unconjugated drug, but it has found clinical success by linkage to antibodies. The anti- CD30 antibody- auristatin conjugate has been approved for cancer therapy (brentuximab vedotin), and several others are in various stages of clinical development including PSMA- ADC (Progenies). We designed a peptide which contains both free -SH from cysteine and free -NH2 from lysine (PSMA-Cys-C6-Lys). Cy5.5 was conjugated to the peptide through - NH2 of the peptide and Vc-MMAE was conjugated to the peptide through reaction of -SH with maleimide group on Vc-MMAE. MMAE is linked to PSMA ligand via the lysosomally cleavable dipeptide, valine-citrulline (vc). The final conjugate PSMA-MMAE-Cy5.5 showed binding affinity at IC50=l.9nM compared to ZJ24 (IC 5 o=5.8nM) (Figs. 16-17). In vitro cytotoxicity experiment showed that PSMA-MMAE-Cy5.5 is about 5-fold more potent to kill PSMA -positive PC3pip cells than to kill PSMA-negative PC3flu cells (Fig. 18). In addition, in vivo imaging further demonstrated selective uptake in PSMA-expressing PC3pip tumors (Figs. 19-21). All these data support the selective targeting ability of PSMA-MMAE-Cy5.5.

[00143] All references cited herein, including patents, patent applications, papers, text books, and the like, and the references cited therein, to the extent that they are not already, are hereby incorporated herein by reference in their entirety. The foregoing description and Examples detail certain preferred embodiments of the invention and describes the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the invention may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof.