Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NTSR1-TARGETED RADIOPHARMACEUTICALS AND DNA DAMAGE RESPONSE INHIBITOR COMBINATION THERAPY
Document Type and Number:
WIPO Patent Application WO/2023/141717
Kind Code:
A1
Abstract:
Methods for treating or ameliorating cancer comprising administering to a mammal a NTSR-1 targeted radiopharmaceutical comprising a radionuclide chelated to a compound of formula I, and DNA damage response inhibitors.

Inventors:
FORBES JOHN R (CA)
MAHAMMAD SALEEMULLA (CA)
Application Number:
PCT/CA2023/050108
Publication Date:
August 03, 2023
Filing Date:
January 27, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FUSION PHARMACEUTICALS INC (CA)
International Classes:
A61K51/04; A61K31/444; A61K31/502; A61K31/519; A61K31/5377; A61P35/00
Domestic Patent References:
WO2014096499A12014-06-26
WO2020115548A12020-06-11
Attorney, Agent or Firm:
SMART & BIGGAR LP (CA)
Download PDF:
Claims:
CLAIMS A method for treating or ameliorating cancer, said method comprising:

(i) administering to a mammal a radiopharmaceutical, wherein the mammal has received or is receiving a DNA damage response inhibitor (DDRi);

(ii) administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical; or

(iii) administering to a mammal a DDRi at the same time as administering to the mammal a radiopharmaceutical, wherein in each occurrence said radiopharmaceutical comprises a radionuclide chelated with a compound of Formula I: wherein

R1 is selected from the group consisting of hydrogen, methyl, and cyclopropylmethyl; AA-COOH is an amino acid selected from the group consisting of 2-amino-2- adamantane carboxylic acid, cyclohexylglycine, and 9-amino-bicyclo[3.3.1]nonane-9- carboxylic acid;

R2 is selected from the group consisting of (Ci-Ce)alkyl, (C3-Cs)cycloalkyl, (C3- Cs)cycloalkylmethyl, halogen, nitro, and trifluoromethyl;

R3 and R4 are each and independently selected from the group consisting of hydrogen and (Ci-C4)alkyl;

Li is (C2-Cs)alkyhdene;

L2 is (C2-C2o)alkylidene, (C2-C2o)heteroalkylidene, (C=O)O, (C=O)NR, or a combination thereof, R being hydrogen or (Ci-C4)alkyl; and

W is a chelator selected from the group consisting of DOTA, DOTAGA, NOTA, DTP A, TETA, EDTA, NOD AGA, NODASA, TRITA, CDTA, BAT, DFO, and HYNIC, wherein the radionuclide is selected from the group consisting of 64Cu, 67Cu, 68Ga, 90Y, i49Tb, i53Sm 177LU, 211At, 212Bi, 212Pb, 213Bi, 223Ra, 225 Ac, and 227Th. The method of claim 1, said method comprising administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical. The method of claim 1 or 2, wherein said radiopharmaceutical is an 225 Ac- radiopharmaceutical comprising 225 Ac chelated with the following structure: The method of any one of claims 1-3, wherein the DDRi is a PARP inhibitor. The method of claim 4, wherein the PARP inhibitor is a small molecule PARP inhibitor. The method of claim 5, wherein the small molecule PARP inhibitor is selected from the group consisting of niparib, niraparib, olaparib, talazoparib, pamiparib, rucaparib (camsylate), and veliparib, or an analog thereof. The method of claim 6, wherein the small molecule PARP inhibitor is olaparib or an analog thereof. The method of any one of claims 1-3, wherein the DDRi is an ATR or ATM inhibitor. The method of claim 8, wherein the ATR or ATM inhibitor is a small molecule ATR or ATM inhibitor. The method of claim 9, wherein the small molecule ATR or ATM inhibitor is selected from the group consisting of AZ20, AZD0156, AZD1390, AZD6738, BAY-1895344, EPT-46464, M3541, M4344, M6620 (formerly known as VE-922 or VX-970), NU6027, and VE-821, or an analog thereof. The method of claim 10, wherein the small molecule ATR or ATM inhibitor is AZD1390, BAY-1895344, or an analog thereof. The method of any one of claims 1-3, wherein the DDRi is a DNA-protein kinase (DNA-PK) inhibitor, a WEE1 inhibitor, a Chkl inhibitor, or a Chk2 inhibitor. The method of claim 12, wherein the DDRi is a DNA-PK inhibitor selected from the group consisting of AZD7648, KU-0060648, NU7026, NU7441 (KU-57788), PI-103, P IK-75 HCI, PP121, and SF2523, or an analog thereof. The method of claim 13, wherein the DNA-PK inhibitor is AZD7648 or an analog thereof. The method of any one of the preceding claims, wherein the mammal is a human. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered at a dosage of less than 1 MBq/kg of body weight of said mammal. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered at a dosage of less than 250 kBq/kg of body weight of said mammal. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered at a dosage of less than 100 kBq/kg of body weight of said mammal. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 15 MBq to said mammal. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 10 MBq to said mammal. The method of any one of claims 3-15, wherein said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 5 MBq to said mammal. The method of any one of the preceding claims, wherein the cancer is selected from the group consisting of colorectal cancer, ductal pancreatic adenocarcinoma, non-small cell lung cancer, small cell lung cancer, prostate cancer, breast cancer, meningioma, Ewing’s sarcoma, pleural mesothelioma, head and neck cancer, gastrointestinal stromal tumors, uterine leiomyoma, sarcoma, adrenocortical carcinoma, neuroendocrine cancer, multiple myeloma, acute myeloid leukemia, and cutaneous T-cell lymphoma . The method of claim 22, wherein said cancer is colorectal cancer or ductal pancreatic adenocarcinoma. The method of any one of the preceding claims, wherein said administering results in a decrease in tumor volume, a stable tumor volume, or a reduced rate of increase in tumor volume. The method of claim 24, wherein said administering results in a decreased incidence of recurrence or metastasis. The method of claim 1, said method comprising administering to a mammal a DDRi, wherein the mammal has received or is receiving an 225 Ac-radiopharmaceutical comprising 225 Ac chelated with the following structure: wherein the DDRi is a PARP inhibitor or an ATR or ATM inhibitor, and wherein said 225 Ac-radiopharmaceutical is administered at a dosage of 100-600 kBq/kg of body weight of said mammal.

Description:
NTSR1-TARGETED RADIOPHARMACEUTICALS AND DNA DAMAGE RESPONSE INHIBITOR COMBINATION THERAPY

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Patent Application No. 63/304,178, filed on January 28, 2022, the entire content of which is incorporated by reference herein.

BACKGROUND

DNA single stranded breaks and double stranded breaks occur for a variety of reasons, including cellular exposure to exogenous sources of DNA damaging agents such as radiopharmaceuticals or due to genetic mutations in the pathways that include the BRCA, PTEN and ATR proteins. Such DNA breaks are repaired through multiple pathways and inhibition of these repair pathways results in an accumulation of single and/or double stranded breaks (e.g., PARP inhibition (PARPi) or ATM inhibition).

Existing PARP inhibitors act through both inhibitors of PARP enzyme inhibition activity and through the trapping of PARP proteins inside of chromatin (“DNA-trapping”). Tumor cells with BRCA and/or PTEN mutations are sensitive to PARPi’s, while ATR inhibition (ATRi) results in a failure to repair double stranded breaks — and therefore leads to the accumulation of double stranded breaks. Likewise, inhibitors of ATM (ATMi) or DNA- PK (DNA-PKi) or other DNA repair pathways can lead to increased accumulation of DNA damage in cells. An increase in single or double stranded DNA breaks in the tumor results in higher cell death. DNA Damage Repair inhibitors (DDRis) have been investigated as cancer therapeutics based on this mechanism. However, the presence of mutations that enable DDRi monotherapy in cancer cells, and which are also found in non-cancerous somatic cells, can result in undesirable normal tissue toxicities. Further, many DDRis have exhibited only modest efficacy in vivo when used as monotherapies and their use may be restricted to cancer types that are already deficient in some aspect of DNA repair capacity (e.g., PARPi for treatment of BRCA1/2 deficient cancers).

Therefore, there is a need for improved treatments of cancer. In particular, there is a need for increases in efficacy, which do not enhance toxicity in the patient.

SUMMARY The present disclosure encompasses the insight that inhibition of DNA damage repair mechanisms used in combination with a therapy that targets DNA breaks specifically to cancer cells (and not normal tissues) may lead to better therapies with improved efficacy. Radioactive decay can cause direct physical damage (such as single or double-stranded DNA breaks) or indirect damage (such as by-stander or crossfire effects) to the biomolecules that constitute a cell. Drugs that deliver radioisotopes, radiopharmaceuticals, to cancer cells provide a mechanism to generate DNA damage with anti-cancer therapeutic effect. The present disclosure provides methods of combining radiopharmaceuticals, specifically, small molecule-based radiopharmaceuticals targeting neurotensin receptor 1 (NTSR1) positive tumors and using actinium-225, lutetium-177 or other suitable radionuclides to target cancer cells, with DDRi to treat or ameliorate cancer.

More specifically, provided are methods of treating or ameliorating cancer, said method comprising:

(i) administering to a mammal a radiopharmaceutical, wherein the mammal has received or is receiving a DNA damage response inhibitor (DDRi);

(ii) administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical; or

(iii) administering to a mammal a DDRi at the same time as administering to the mammal a radiopharmaceutical, wherein in each occurrence said radiopharmaceutical comprises a radionuclide chelated with a compound of Formula I: wherein

R 1 is selected from the group consisting of hydrogen, methyl, and cyclopropylmethyl; AA-COOH is an amino acid selected from the group consisting of 2-amino-2- adamantane carboxylic acid, cyclohexylglycine, and 9-amino-bicyclo[3.3.1]nonane-9- carboxylic acid;

R 2 is selected from the group consisting of Ci-6 alkyl, C3-8 cycloalkyl, C3-8 cycloalkylmethyl, halogen, nitro, and trifluoromethyl;

R 3 and R 4 are each independently selected from the group consisting of hydrogen and Ci -4 alkyl;

Li is C2-5 alkylidene;

L2 is C2-20 alkylidene, C2-20 heteroalkylidene, (C=O)O, (C=O)NR, or a combination thereof, R being hydrogen or C 1-4 alkyl; and

W is a chelator selected from the group consisting of DOT A, DOTAGA, NOTA, DTP A, TETA, EDTA, NOD AGA, NODASA, TRITA, CDTA, BAT, DFO, and HYNIC, wherein the radionuclide is selected from the group consisting of 64 Cu, 67 Cu, 68 Ga, 90 Y, i49 Tb, i53 Sm 177 LU , 211 At, 212 Bi, 212 Pb, 213 Bi, 223 Ra, 225 Ac, and 227 Th.

In some embodiments, said method comprising administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical.

In some embodiments, said method comprises administering to a mammal a radiopharmaceutical, wherein the mammal has received or is receiving one or more DDRi.

In some embodiments, said method comprises administering to a mammal one or more DDRi at the same time as administering to the mammal a radiopharmaceutical.

In some embodiments, the chelator is selected from the group consisting of DOTA, DOTA-GA, NOTA, NODA-GA, and NODA-SA.

In some embodiments, the chelator is selected from the group consisting of DTP A, EDTA, CDTA, DFO, BAT, and HYNIC.

In some embodiments, said radiopharmaceutical is an 225 Ac-radiopharmaceutical comprising 225 Ac chelated with a compound of Formula I. An exemplary 225 Ac- radiopharmaceutical comprises 225 Ac chelated with the following structure (Compound A): Compound A

In some embodiments, the DDRi is a PARP inhibitor. In certain embodiments, the PARP inhibitor is a small molecule PARP inhibitor. In certain embodiments, the small molecule PARP inhibitor is selected from the group consisting of niparib, niraparib, olaparib, talazoparib, pamiparib, rucaparib (camsylate), and veliparib, or an analog thereof. In certain embodiments, the small molecule PARP inhibitor is olaparib or an analog thereof.

In some embodiments, the DDRi is an ATR or ATM inhibitor. In certain embodiments, the ATR or ATM inhibitor is a small molecule ATR or ATM inhibitor. In certain embodiments, the small molecule ATR or ATM inhibitor is selected from the group consisting of AZ20, AZD0156, AZD1390, AZD6738, BAY-1895344, EPT-46464, M3541, M4344, M6620 (formerly known as VE-922 or VX-970), NU6027, and VE-821, or an analog thereof. In certain embodiments, the small molecule ATR or ATM inhibitor is AZDI 390, BAY-1895344, or an analog thereof.

In some embodiments, the DDRi is a DNA-protein kinase (DNA-PK) inhibitor, a WEE1 inhibitor, a Chkl inhibitor, or a Chk2 inhibitor. In certain embodiments, the DDRi is a DNA-PK inhibitor selected from the group consisting of AZD7648, KU-0060648, NU7026, NU7441 (KU-57788), PI-103, PIK-75 HCI, PP121, and SF2523, or an analog thereof. In certain embodiments, the DNA-PK inhibitor is AZD7648 or an analog thereof.

In some embodiments, the mammal is a human.

In some embodiments, said 225 Ac-radiopharmaceutical is administered at a dosage of less than 1 MBq/kg of body weight of said mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered at a dosage of less than 250 kBq/kg of body weight of said mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered at a dosage of less than 100 kBq/kg of body weight of said mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 15 MBq to said mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 10 MBq to said mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 5 MBq to said mammal.

In some embodiments, said DDRi is administered at a dosage of about 5 mg/kg to about 30 mg/kg of body weight of said mammal. In some embodiments, said DDRi is administered at a dosage of about 25 mg/kg of body weight of said mammal.

In some embodiments, the cancer is selected from the group consisting of colorectal cancer, ductal pancreatic adenocarcinoma, non-small cell lung cancer, small cell lung cancer, prostate cancer, breast cancer, meningioma, Ewing’s sarcoma, pleural mesothelioma, head and neck cancer, gastrointestinal stromal tumors, uterine leiomyoma, sarcoma, adrenocortical carcinoma, neuroendocrine can cer, multiple myeloma, acute myeloid leukemia, and cutaneous T-cell lymphoma .

In some embodiments, said cancer is colorectal cancer or ductal pancreatic adenocarcinoma.

In some embodiments, said administration results in a decrease in tumor volume, a stable tumor volume, or a reduced rate of increase in tumor volume.

In some embodiments, said administration results in a decreased incidence of recurrence or metastasis.

In some embodiments, said method comprising administering to a mammal a DDRi, wherein the mammal has received or is receiving an 225 Ac-radiopharmaceutical comprising 225 Ac chelated with the following structure: wherein the DDRi is a PARP inhibitor or an ATR or ATM inhibitor, and wherein said 225 Ac-radiopharmaceutical is administered at a dosage of 100-600 kBq/kg of body weight of said mammal.

Also provided herein is a use of a compound of Formula I for the manufacture of a medicament for a method of treating or ameliorating cancer in a subject in need thereof, said method comprising:

(i) administering to a mammal a radiopharmaceutical, wherein the mammal has received or is receiving a DNA damage response inhibitor (DDRi); (ii) administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical; or

(iii) administering to a mammal a DDRi at the same time as administering to the mammal a radiopharmaceutical, wherein the compound of Formula I is represented by: wherein

R 1 is selected from the group consisting of hydrogen, methyl, and cyclopropylmethyl;

AA-COOH is an amino acid selected from the group consisting of 2-amino-2- adamantane carboxylic acid, cyclohexylglycine, and 9-amino-bicyclo[3.3.1]nonane-9- carboxylic acid;

R 2 is selected from the group consisting of Ci-6 alkyl, C3-8 cycloalkyl, C3-8 cycloalkylmethyl, halogen, nitro, and trifluoromethyl;

R 3 and R 4 are each independently selected from the group consisting of hydrogen and Ci -4 alkyl;

Li is C2-5 alkylidene;

L2 is C2-20 alkylidene, C2-20 heteroalkylidene, (C=O)O, (C=O)NR, or a combination thereof, R being hydrogen or C 1-4 alkyl; and

W is a chelator selected from the group consisting of DOT A, DOTAGA, NOTA, DTP A, TETA, EDTA, NOD AGA, NODASA, TRITA, CDTA, BAT, DFO, and HYNIC, wherein the radionuclide is selected from the group consisting of 64 Cu, 67 Cu, 68 Ga, 90 Y, i49 Tb , i53 Sm 177 LU , 211 At, 212 Bi, 212 Pb, 213 Bi, 223 Ra, 225 Ac, and 227 Th; and wherein in each occurrence said radiopharmaceutical comprises a radionuclide chelated with the compound of Formula I. In another aspect, provided herein is a compound of Formula I for use in treating or ameliorating cancer in a subject in need thereof, said use comprising:

(i) administering to a mammal a radiopharmaceutical, wherein the mammal has received or is receiving a DNA damage response inhibitor (DDRi);

(ii) administering to a mammal a DDRi, wherein the mammal has received or is receiving a radiopharmaceutical; or

(iii) administering to a mammal a DDRi at the same time as administering to the mammal a radiopharmaceutical, wherein the compound of Formula I is represented by: wherein

R 1 is selected from the group consisting of hydrogen, methyl, and cyclopropylmethyl;

AA-COOH is an amino acid selected from the group consisting of 2-amino-2- adamantane carboxylic acid, cyclohexylglycine, and 9-amino-bicyclo[3.3.1]nonane-9- carboxylic acid;

R 2 is selected from the group consisting of Ci-6 alkyl, C3-8 cycloalkyl, C3-8 cycloalkylmethyl, halogen, nitro, and trifluoromethyl;

R 3 and R 4 are each independently selected from the group consisting of hydrogen and Ci -4 alkyl;

Li is C2-5 alkylidene;

L2 is C2-20 alkylidene, C2-20 heteroalkylidene, (C=O)O, (C=O)NR, or a combination thereof, R being hydrogen or C 1-4 alkyl; and

W is a chelator selected from the group consisting of DOT A, DOTAGA, NOTA, DTP A, TETA, EDTA, NOD AGA, NODASA, TRITA, CDTA, BAT, DFO, and HYNIC; and wherein in each occurrence said radiopharmaceutical comprises a radionuclide chelated with the compound of Formula I, wherein the radionuclide is selected from the group consisting of 64 Cu, 67 Cu, 68 Ga, 90 Y, 149 Tb, 153 Sm, 177 Lu, 211 At, 212 Bi, 212 Pb, 213 Bi, 223 Ra, 225 Ac, and 227 Th.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 illustrates the biodistribution of radiopharmaceutical [ 177 Lu] -Compound A in a CT-26-mNTSRl syngeneic immunocompetent mouse model.

Figure 2 illustrates the in vivo efficacy of radiopharmaceutical [ 225 Ac] -Compound A at different dosages in a CT-26-mNTSRl xenograft model.

Figure 3 illustrates increased therapeutic efficacy from the combination of radiopharmaceutical [ 225 Ac] -Compound A and Olaparib in a CT-26-mNTSRl xenograft model.

Figure 4 illustrates improvement of the overall survival in mice treated with a combination of [ 225 Ac] -Compound A and olaparib.

DETAILED DESCRIPTION

The present disclosure relates to combination therapies for treating or ameliorating cancer using certain radiopharmaceuticals and DNA damage response inhibitors in combination. In particular, the radiopharmaceuticals are radionuclide-chelated small molecules targeting neurotensin receptor 1 (NTSR1).

NTSR1 is a transmembrane receptor that binds the neurotransmitter neurotensin (Vincent el al., Trends Pharmacol. Sci., 1999, 20, 302-309; Pelaprat, Pepti des, 2006, 27, 2476-2487). NTSR1 is expressed predominantly in the central nervous system and intestine (smooth muscle, mucosa and nerve cells). Apart from the central nervous system, NTSR1 is highly expressed in a mammalian body and a human body in particular on several neoplastic cells in several tumor indications, whereas the expression of NTSR1 in most other tissues of the mammalian and the human body is either not existent or low. See, e.g., Bugni et al., Ini. J. Cancer, 2012, 730,1798-1805; Wang et al., Neuropeptides, 2011, 45, 151-156; and Taylor et al., Prostate, 2012, 72, 523-32.

These NTSR1 expressing tumor indications include but are not limited to ductal pancreatic adenocarcinoma, small cell lung cancer, prostate cancer, colorectal cancer, breast cancer, meningioma, Ewing’s sarcoma, pleural mesothelioma, head and neck cancer, nonsmall cell lung cancer, gastrointestinal stromal tumors, uterine leiomyoma, and cutaneous T- cell lymphoma. A preferred group of NTSR1 expressing tumor indications are ductal pancreatic adenocarcinoma, small cell lung cancer, prostate cancer, colorectal cancer, breast cancer, meningioma, and Ewing’s sarcoma.

Radio-labelled targeting moieties (also known as radiopharmaceuticals) are designed to target a protein or receptor (e.g., NTSR1) that is upregulated in a disease state and/or specific to diseased cells (e.g., tumor cells) to deliver a radioactive payload to damage and kill cells of interest.

Definitions

Chemical Terms

The term “alkyl,” as used herein, is inclusive of both straight chain and branched chain saturated groups from 1 to 20 carbons (e.g., from 1 to 10 or from 1 to 6), unless otherwise specified. Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, neopentyl, and the like, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents independently selected from the group consisting of: (1) Ci-6 alkoxy; (2) Ci-6 alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(R N1 )2, where R N1 is as defined for amino); (4) Ce-io aryl-Ci-6 alkoxy; (5) azido; (6) halo; (7) (C2-9 heterocyclyl)oxy; (8) hydroxy, optionally substituted with an (9-protecting group; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) C1-7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) -CO2R A , optionally substituted with an (9-protecting group and where R A is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce-io aryl, (d) hydrogen, (e) C1-6 alk-Ce-io aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci- 20 alkyl, and (h) amino-polyethylene glycol of -NR N1 (CH2)s2(CH2CH2O) s i(CH2)s3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (15) -C(O)NR B R c , where each of R B and R c is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (16) -SO2R D , where R D is selected from the group consisting of (a) C1-6 alkyl, (b) Ce-io aryl, (c) C1-6 alk-Ce-io aryl, and (d) hydroxy; (17) -SO2NR E R F , where each of R E and R F is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl and (d) Ci-6 alk-Ce-io aryl; (18) -C(O)R G , where R G is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce-io aryl, (d) hydrogen, (e) C1-6 alk-Ce-io aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of - (CH2)s2(OCH2CH2)si(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C1-20 alkyl, and (h) amino-polyethylene glycol of -NR N1 (CH2)S2(CH2CH2O) S I(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (19) - NR H C(O)R J , wherein R H is selected from the group consisting of (al) hydrogen and (bl) Ci- 6 alkyl, and R 1 is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) Ce-io aryl, (d2) hydrogen, (e2) C1-6 alk-Ce-io aryl, (f2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2)si(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci -20 alkyl, and (h2) amino-polyethylene glycol of - NR N1 (CH2)S2(CH 2 CH 2 O)SI(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (20) -NR J C(O)OR K , wherein R J is selected from the group consisting of (al) hydrogen and (bl) C1-6 alkyl, and R K is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) Ce-io aryl, (d2) hydrogen, (e2) C1-6 alk-Ce-io aryl, (£2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci- 20 alkyl, and (h2) amino-polyethylene glycol of -NR N1 (CH2)S2(CH2CH2O) S I(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl can be further substituted with an oxo group to afford the respective aryloyl substituent. The terms “alkylene”, “alkylidene”, and the prefix “alk-,” as used herein, represent a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The terms “C x-y alkyl,” “C x-y alkylene,” “C x.y alkylidene,” and the prefix “C x-y alk-” represent alkyl or alkylene groups having between x and y carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., Ci-6, Ci-io, C2-5, C2-8, C2-10, or C2-20 alkyl or alkylene). In some embodiments, the alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for an alkyl group.

The term “alkenyl,” as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1 -propenyl, 2-propenyl, 2 -methyl- 1 -propenyl, 1-butenyl, 2-butenyl, and the like. Alkenyls include both cis and trans isomers. Alkenyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from amino, aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.

The term “alkynyl,” as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, 1-propynyl, and the like. Alkynyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.

The term “amino,” as used herein, represents -N(R N1 )2, wherein each R N1 is, independently, H, OH, NO2, N(R N2 )2, SO2OR N2 , SO2R N2 , SOR N2 , an ^-protecting group, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, carboxyalkyl (e.g., optionally substituted with an (9-protecting group, such as optionally substituted arylalkoxycarbonyl groups or any described herein), sulfoalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), alkoxy carbonylalkyl (e.g., optionally substituted with an (9-protecting group, such as optionally substituted arylalkoxy carbonyl groups or any described herein), heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), wherein each of these recited R N1 groups can be optionally substituted, as defined herein for each group; or two R N1 combine to form a heterocyclyl or an ^-protecting group, and wherein each R N2 is, independently, H, alkyl, or aryl. Amino groups can be unsubstituted amino (i.e., -NH2) or substituted amino (i.e., -N(R N1 )2) groups. In a preferred embodiment, amino is -NH2 or -NHR N1 , wherein R N1 is, independently, OH, NO2, NH2, NR N2 2, SO2OR N2 , SO2R N2 , SOR N2 , alkyl, carboxyalkyl, sulfoalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), alkoxy carbonylalkyl (e.g., t-butoxy carbonylalkyl) or aryl, and each R N2 can be H, C1-20 alkyl (e.g., C1-6 alkyl), or Ce-io aryl.

The term “amino acid,” as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., a carboxy group of-CO2H or a sulfo group of-SOsH), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). In some embodiments, the amino acid is attached to the parent molecular group by a carbonyl group, where the side chain or amino group is attached to the carbonyl group. Exemplary side chains include an optionally substituted alkyl, aryl, heterocyclyl, alkaryl, alkheterocyclyl, aminoalkyl, carbamoylalkyl, and carboxyalkyl. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, hydroxynorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine, phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine. Amino acid groups may be optionally substituted with one, two, three, or, in the case of amino acid groups of two carbons or more, four substituents independently selected from the group consisting of: (1) C1-6 alkoxy; (2) C1-6 alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(R N1 )2, where R N1 is as defined for amino); (4) Ce- 10 aryl-Ci-6 alkoxy; (5) azido; (6) halo; (7) (C2-9heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo (e.g., carboxy aldehyde or acyl); (11) C1-7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) - CO2R A , where R A is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce-io aryl, (d) hydrogen, (e) C1-6 alk-Ce-io aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci -20 alkyl, and (h) amino-polyethylene glycol of - NR N1 (CH2)S2(CH 2 CH 2 O)SI(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (15) -C(O)NR B R c , where each of R B and R c is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (16) -SO2R D , where R D is selected from the group consisting of (a) Ci-6 alkyl, (b) Ce-io aryl, (c) Ci-6 alk-Ce-io aryl, and (d) hydroxy; (17) -SO2NR E R F , where each of R E and R F is, independently, selected from the group consisting of (a) hydrogen, (b) Ci-6 alkyl, (c) Ce-io aryl and (d) Ci-6 alk-Ce-io aryl; (18) -C(O)R G , where R G is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) Ce-io aryl, (d) hydrogen, (e) C1-6 alk-Ce-io aryl, (f) amino-Ci-20 alkyl, (g) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci- 20 alkyl, and (h) amino-polyethylene glycol of -NR N1 (CH2)S2(CH2CH2O) S I(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (19) -NR H C(O)R J , wherein R H is selected from the group consisting of (al) hydrogen and (bl) C1-6 alkyl, and R 1 is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) Ce-io aryl, (d2) hydrogen, (e2) C1-6 alk-Ce-io aryl, (f2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or C1-20 alkyl, and (h2) amino-polyethylene glycol of - NR N1 (CH 2 )S2(CH2CH2O)S1(CH 2 )S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (20) -NR J C(O)OR K , wherein R J is selected from the group consisting of (al) hydrogen and (bl) C1-6 alkyl, and R K is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) Ce-io aryl, (d2) hydrogen, (e2) C1-6 alk-Ce-io aryl, (£2) amino-Ci-20 alkyl, (g2) polyethylene glycol of -(CH2)s2(OCH2CH2) s i(CH2)s3OR’, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R’ is H or Ci- 20 alkyl, and (h2) amino-polyethylene glycol of -NR N1 (CH2)S2(CH2CH2O) S I(CH2)S3NR N1 , wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted C1-6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein.

The term “aryl,” as used herein, represents a mono-, bicyclic, or multicyclic carbocyclic ring system having one or two aromatic rings and is exemplified by phenyl, naphthyl, 1,2- dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, phenanthrenyl, fluorenyl, indanyl, indenyl, and the like, and may be optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from the group consisting of: (1) C1-7 acyl (e.g., carboxy aldehyde); (2) C1-20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-Ci-6 alkyl, C1-6 alkylsulfinyl-Ci-6 alkyl, amino-Ci-6 alkyl, azido-Ci-6 alkyl, (carboxyaldehyde)-Ci-6 alkyl, halo-Ci-6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci-6 alkyl, nitro-Ci-6 alkyl, or C1-6 thioalkoxy-Ci-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) Ce-io aryl; (6) amino; (7) C1-6 alk- Ce-io aryl; (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-Cs-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) -(CH2) q CO2R A , where q is an integer from zero to four, and R A is selected from the group consisting of (a) C1-6 alkyl, (b) Ce-io aryl, (c) hydrogen, and (d) C1-6 alk-Ce-io aryl; (18) -(CH2) q CONR B R c , where q is an integer from zero to four and where R B and R c are independently selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (19) -(CH2) q SO2R D , where q is an integer from zero to four and where R D is selected from the group consisting of (a) alkyl, (b) Ce-io aryl, and (c) alk-Ce-io aryl; (20) -(CH2) q SO2NR E R F , where q is an integer from zero to four and where each of R E and R F is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (21) thiol; (22) Ce-io aryloxy; (23) C3-8 cycloalkoxy; (24) Ce-io aryl-Ci-6 alkoxy; (25) C1-6 alk-Ci-12 heterocyclyl (e.g., C1-6 alk-Ci-12 heteroaryl); (26) C2-20 alkenyl; and (27) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci-alkheterocyclylcan be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “arylalkyl,” as used herein, represents an aryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted arylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-6 alk-Ce-io aryl, C1-10 alk-Ce-io aryl, or C1-20 alk-Ce-io aryl). In some embodiments, the alkylene and the aryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups. Other groups preceded by the prefix “alk-” are defined in the same manner, where “alk” refers to a Ci-6 alkylene, unless otherwise noted, and the attached chemical structure is as defined herein.

The term “carbonyl,” as used herein, represents a C(O) group, which can also be represented as C=O.

The term “carboxy,” as used herein, means -CO2H.

The term “cyano,” as used herein, represents an -CN group.

The term “cycloalkyl,” as used herein represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicycle heptyl, and the like. When the cycloalkyl group includes one carboncarbon double bond or one carbon-carbon triple bond, the cycloalkyl group can be referred to as a “cycloalkenyl” or “cycloalkynyl” group respectively. Exemplary cycloalkenyl and cycloalkynyl groups include cyclopentenyl, cyclohexenyl, cyclohexynyl, and the like. Cycloalkyl groups can be optionally substituted with: (1) C1-7 acyl (e.g., carboxyaldehyde); (2) Ci -20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-Ci-6 alkyl, C1-6 alkylsulfinyl-Ci-6 alkyl, amino-Ci-6 alkyl, azido-Ci-6 alkyl, (carboxyaldehyde)-Ci-6 alkyl, halo-Ci-6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci-6 alkyl, nitro-Ci-6 alkyl, or Ci-6 thioalkoxy-Ci-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) Ce-io aryl; (6) amino; (7) C1-6 alk- Ce-io aryl; (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-Cs-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) -(CH2) q CO2R A , where q is an integer from zero to four, and R A is selected from the group consisting of (a) C1-6 alkyl, (b) Ce-io aryl, (c) hydrogen, and (d) C1-6 alk-Ce-io aryl; (18) -(CH2) q CONR B R c , where q is an integer from zero to four and where R B and R c are independently selected from the group consisting of (a) hydrogen, (b) Ce-io alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (19) -(CH2) q SO2R D , where q is an integer from zero to four and where R D is selected from the group consisting of (a) Ce-io alkyl, (b) Ce-io aryl, and (c) C1-6 alk-Ce-io aryl; (20) -(CH2) q SO2NR E R F , where q is an integer from zero to four and where each of R E and R F is, independently, selected from the group consisting of (a) hydrogen, (b) Ce-io alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (21) thiol; (22) Ce-io aryloxy; (23) C3-8 cycloalkoxy; (24) Ce-io aryl-Ci-6 alkoxy; (25) C1-6 alk- C1-12 heterocyclyl (e.g., C1-6 alk-Ci-12 heteroaryl); (26) oxo; (27) C2-20 alkenyl; and (28) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “diastereomer,” as used herein means stereoisomers that are not mirror images of one another and are non-superimposable on one another.

The term “enantiomer,” as used herein, means each individual optically active form of a compound, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.

The term “halogen,” as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.

The terms “heteroalkyl” and “heteroalkylidene,” as used herein, each refer to an alkyl group, as defined herein, in which one or two of the constituent carbon atoms have each been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups. The terms “heteroalkenyl” and heteroalkynyl,” as used herein refer to alkenyl and alkynyl groups, as defined herein, respectively, in which one or two of the constituent carbon atoms have each been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkenyl and heteroalkynyl groups can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.

The term “heteroaryl,” as used herein, represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4/?+2 pi electrons within the mono- or multi cyclic ring system. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. In some embodiment, the heteroaryl is substituted with 1, 2, 3, or 4 substituents groups as defined for a heterocyclyl group.

The term “heteroarylalkyl” refers to a heteroaryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted heteroarylalkyl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as Ci-6 alk-Ci-12 heteroaryl, C1-10 alk-Ci-12 heteroaryl, or C1-20 alk-Ci-12 heteroaryl). In some embodiments, the alkylene and the heteroaryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group. Heteroarylalkyl groups are a subset of heterocyclylalkyl groups. The term “heterocyclyl,” as used herein represents a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5 -membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocyclyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. The term “heterocyclyl” also represents a heterocyclic compound having a bridged multi cyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocyclyl” includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like. Examples of fused heterocyclyls include tropanes and l,2,3,5,8,8a-hexahydroindolizine. Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, dihydroquinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzothiadiazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl (e.g., 1,2,3-oxadiazolyl), purinyl, thiadiazolyl (e.g., 1,2,3-thiadiazolyl), tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl, dihydroquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydroisoquinolyl, pyranyl, dihydropyranyl, dithiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, and the like, including dihydro and tetrahydro forms thereof, where one or more double bonds are reduced and replaced with hydrogens. Still other exemplary heterocyclyls include: 2,3,4,5-tetrahydro-2-oxo-oxazolyl; 2,3-dihydro-2-oxo-lH-imidazolyl; 2,3,4,5-tetrahydro-5-oxo-lH-pyrazolyl (e.g., 2,3,4,5-tetrahydro-2-phenyl-5-oxo-lH- pyrazolyl); 2,3,4,5-tetrahydro-2,4-dioxo-lH-imidazolyl (e.g., 2,3,4,5-tetrahydro-2,4-dioxo-5- methyl-5-phenyl-lH-imidazolyl); 2,3-dihydro-2-thioxo-l,3,4-oxadiazolyl (e.g., 2,3-dihydro- 2-thioxo-5-phenyl-l,3,4-oxadiazolyl); 4,5-dihydro-5-oxo-17/-triazolyl (e.g., 4,5-dihydro-3- methyl-4-amino 5-oxo-l/f-triazolyl); l,2,3,4-tetrahydro-2,4-dioxopyridinyl (e.g., 1, 2,3,4- tetrahydro-2,4-dioxo-3,3-diethylpyridinyl); 2,6-dioxo-piperidinyl (e.g., 2,6-dioxo-3-ethyl-3- phenylpiperidinyl); l,6-dihydro-6-oxopyridiminyl; l,6-dihydro-4-oxopyrimidinyl (e.g., 2- (methylthio)- l,6-dihydro-4-oxo-5-methylpyrimi din- 1-yl); l,2,3,4-tetrahydro-2,4- dioxopyrimidinyl (e.g., l,2,3,4-tetrahydro-2,4-dioxo-3-ethylpyrimidinyl); l,6-dihydro-6-oxo- pyridazinyl (e.g., l,6-dihydro-6-oxo-3-ethylpyridazinyl); l,6-dihydro-6-oxo-l,2,4-triazinyl (e.g., l,6-dihydro-5-isopropyl-6-oxo-l,2,4-triazinyl); 2,3-dihydro-2-oxo-17/-indolyl (e.g., 3,3-dimethyl-2,3-dihydro-2-oxo-17/-indolyl and 2,3-dihydro-2-oxo-3,3'-spiropropane-17/- indol-l-yl); l,3-dihydro-l-oxo-27/-iso-indolyl; l,3-dihydro-l,3-dioxo-27/-iso-indolyl; 1H- benzopyrazolyl (e.g., 1 -(ethoxy carbonyl)- 1/7-benzopyrazolyl); 2,3-dihydro-2-oxo-17/- benzimidazolyl (e.g., 3-ethyl-2,3-dihydro-2-oxo-17/-benzimidazolyl); 2,3-dihydro-2-oxo- benzoxazolyl (e.g., 5-chloro-2,3-dihydro-2-oxo-benzoxazolyl); 2,3-dihydro-2-oxo- benzoxazolyl; 2-oxo-2H-benzopyranyl; 1,4-benzodioxanyl; 1,3-benzodioxanyl; 2,3-dihydro- 3-oxo, 47/-l,3-benzothiazinyl; 3,4-dihydro-4-oxo-37/-quinazolinyl (e.g., 2-methyl-3,4- dihydro-4-oxo-37/-quinazolinyl); l,2,3,4-tetrahydro-2,4-dioxo-37/-quinazolyl (e.g., 1-ethyl- l,2,3,4-tetrahydro-2,4-dioxo-37/-quinazolyl); l,2,3,6-tetrahydro-2,6-dioxo-77/-purinyl (e.g., l,2,3,6-tetrahydro-l,3-dimethyl-2,6-dioxo-7 Ff -purinyl); l,2,3,6-tetrahydro-2,6-dioxo-l H - purinyl (e.g., l,2,3,6-tetrahydro-3,7-dimethyl-2,6-dioxo-l Ff -purinyl); 2-oxobenz[c,<7]indolyl; l . l-dioxo-2H-naphth| l.8- .t/|isothiazolyl; and 1,8-naphthylenedicarboxamido. Additional heterocyclics include 3,3a,4,5,6,6a-hexahydro-pyrrolo[3,4-b]pyrrol-(2H)-yl, and 2,5- diazabicyclo[2.2.1]heptan-2-yl, homopiperazinyl (or diazepanyl), tetrahydropyranyl, dithiazolyl, benzofuranyl, benzothienyl, oxepanyl, thiepanyl, azocanyl, oxecanyl, and thiocanyl. Heterocyclic groups also include groups of the formula

E' is selected from the group consisting of -N- and -CH-; F' is selected from the group consisting of -N=CH-, -NH-CH 2 -, -NH-C(O)-, -NH-, -CH=N-, -CH 2 -NH-, -C(0)-NH-, - CH=CH-, -CH 2 -, -CH 2 CH 2 -, -CH 2 0-, -0CH 2 -, -0-, and -S-; and G' is selected from the group consisting of -CH- and -N-. Any of the heterocyclyl groups mentioned herein may be optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of: (1) C1-7 acyl (e.g., carboxyaldehyde ); (2) C1-20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-Ci-6 alkyl, C1-6 alkylsulfinyl-Ci-6 alkyl, amino-Ci-6 alkyl, azido-Ci-6 alkyl, (carboxyaldehyde)-Ci-6 alkyl, halo-Ci-6 alkyl (e.g., perfluoroalkyl), hydroxy-Ci-6 alkyl, nitro- C1-6 alkyl, or C1-6 thioalkoxy-Ci-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) Ce-io aryl; (6) amino; (7) C1-6 alk-Ce-io aryl; (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-Cs-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C2-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) -(CH2) q CO2R A , where q is an integer from zero to four, and R A is selected from the group consisting of (a) C1-6 alkyl, (b) Ce-io aryl, (c) hydrogen, and (d) C1-6 alk-Ce-io aryl; (18) -(CH2) q C0NR B R c , where q is an integer from zero to four and where R B and R c are independently selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (19) -(CH2) q SO2R D , where q is an integer from zero to four and where R D is selected from the group consisting of (a) C1-6 alkyl, (b) Ce-io aryl, and (c) C1-6 alk-Ce-io aryl; (20) -(CH2) q SO2NR E R F , where q is an integer from zero to four and where each of R E and R F is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) Ce-io aryl, and (d) C1-6 alk-Ce-io aryl; (21) thiol; (22) Ce-io aryloxy; (23) C3-8 cycloalkoxy; (24) arylalkoxy; (25) C1-6 alk-Ci-12 heterocyclyl (e.g., C1-6 alk-Ci-12 heteroaryl); (26) oxo; (27) (C1-12 heterocyclyl)imino; (28) C2- 20 alkenyl; and (29) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a Ci-alkaryl or a Ci- alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “hydrocarbon,” as used herein, represents a group consisting only of carbon and hydrogen atoms.

The term “hydroxyl,” as used herein, represents an -OH group. In some embodiments, the hydroxyl group can be substituted with 1, 2, 3, or 4 substituent groups (e.g., (9-protecting groups) as defined herein for an alkyl.

The term “isomer,” as used herein, means any tautomer, stereoisomer, enantiomer, or diastereomer of any compound. It is recognized that the compounds can have one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (-)) or cis/trans isomers). Unless otherwise noted, chemical structures depicted herein encompass all of the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds can typically be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.

The term ‘W-protected amino,” as used herein, refers to an amino group, as defined herein, to which is attached one or two ^-protecting groups, as defined herein.

The term ‘W-protecting group,” as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used ^-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference, ^-protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4- bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine, and the like; sulfonyl-containing groups such as benzenesulfonyl, p-toluenesulfonyl, and the like; carbamate forming groups such as benzyloxy carbonyl, p-chlorobenzyloxy carbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxy carbonyl, 2 -nitrobenzyloxy carbonyl, p-bromobenzyloxy carbonyl, 3,4- dimethoxybenzyloxy carbonyl, 3, 5-dimethoxybenzyloxy carbonyl, 2,4- dimethoxybenzyloxy carbonyl, 4-methoxybenzyloxy carbonyl, 2-nitro-4,5- dimethoxybenzyloxy carbonyl, 3, 4, 5 -trimethoxy benzyloxy carbonyl, 1 -(p-biphenylyl)-l - methylethoxy carbonyl, a, a-dimethyl-3, 5-dimethoxybenzyloxy carbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxy carbonyl, methoxy carbonyl, allyloxy carbonyl, 2, 2, 2, -trichloroethoxy carbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxy carbonyl, adamantyloxy carbonyl, cyclohexyloxy carbonyl, phenylthiocarbonyl, and the like, alkaryl groups such as benzyl, triphenylmethyl, benzyloxymethyl, and the like and silyl groups, such as trimethylsilyl, and the like. Preferred ^-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).

The term "(□-protecting group,” as used herein, represents those groups intended to protect an oxygen containing (e.g., phenol, hydroxyl, or carbonyl) group against undesirable reactions during synthetic procedures. Commonly used (□-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. Exemplary (□-protecting groups include acyl, aryloyl, or carbamyl groups, such as formyl, acetyl, propionyl, pivaloyl, t- butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o- nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, t- butyldimethylsilyl, tri-/.s -propylsilyloxymethyl. 4,4'-dimethoxytrityl, isobutyryl, phenoxyacetyl, 4-isopropylpehenoxyacetyl, dimethylformamidino, and 4-nitrobenzoyl; alkylcarbonyl groups, such as acyl, acetyl, propionyl, pivaloyl, and the like; optionally substituted arylcarbonyl groups, such as benzoyl; silyl groups, such as trimethylsilyl (TMS), tert-butyldimethylsilyl (TBDMS), tri-iso-propylsilyloxymethyl (TOM), triisopropylsilyl (TIPS), and the like; ether-forming groups with the hydroxyl, such methyl, methoxymethyl, tetrahydropyranyl, benzyl, p-methoxybenzyl, trityl, and the like; alkoxycarbonyls, such as methoxy carbonyl, ethoxy carbonyl, isopropoxycarbonyl, n-isopropoxy carbonyl, n- butyloxy carbonyl, isobutyloxy carbonyl, sec-butyloxy carbonyl, t-butyloxy carbonyl, 2- ethylhexyloxycarbonyl, cyclohexyloxy carbonyl, methyloxycarbonyl, and the like; alkoxyalkoxycarbonyl groups, such as methoxymethoxycarbonyl, ethoxymethoxycarbonyl, 2 -methoxy ethoxy carbonyl, 2-ethoxy ethoxy carbonyl, 2 -butoxy ethoxy carbonyl, 2- methoxyethoxymethoxycarbonyl, allyloxy carbonyl, propargyloxy carbonyl, 2- butenoxy carbonyl, 3-methyl-2-butenoxy carbonyl, and the like; haloalkoxy carbonyls, such as 2-chloroethoxy carbonyl, 2-chloroethoxy carbonyl, 2, 2, 2-trichloroethoxy carbonyl, and the like; optionally substituted arylalkoxycarbonyl groups, such as benzyloxy carbonyl, p- methylbenzyloxy carbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxy carbonyl, 2,4- dinitrobenzyloxycarbonyl, 3,5-dimethylbenzyloxycarbonyl, p-chlorobenzyloxy carbonyl, p- bromobenzyloxy-carbonyl, fluorenylmethyloxycarbonyl, and the like; and optionally substituted aryloxy carbonyl groups, such as phenoxycarbonyl, p-nitrophenoxycarbonyl, o- nitrophenoxy carbonyl, 2, 4-dinitrophenoxy carbonyl, p-methyl-phenoxycarbonyl, m- methylphenoxy carbonyl, o-bromophenoxy carbonyl, 3, 5 -dimethylphenoxy carbonyl, p- chlorophenoxy carbonyl, 2-chloro-4-nitrophenoxy-carbonyl, and the like); substituted alkyl, aryl, and alkaryl ethers (e.g., trityl; methylthiomethyl; methoxy methyl; benzyloxymethyl; siloxymethyl; 2, 2, 2, -trichloroethoxymethyl; tetrahydropyranyl; tetrahydrofuranyl; ethoxyethyl; l-[2-(trimethylsilyl)ethoxy]ethyl; 2-trimethylsilylethyl; t-butyl ether; p- chlorophenyl, p-methoxyphenyl, p-nitrophenyl, benzyl, p-methoxybenzyl, and nitrobenzyl); silyl ethers (e.g., trimethylsilyl; triethylsilyl; triisopropylsilyl; dimethylisopropylsilyl; t- butyldimethylsilyl; t-butyldiphenylsilyl; tribenzylsilyl; triphenylsilyl; and diphenymethylsilyl); carbonates (e.g., methyl, methoxymethyl, 9-fluorenylmethyl; ethyl; 2,2,2-trichloroethyl; 2-(trimethylsilyl)ethyl; vinyl, allyl, nitrophenyl; benzyl; methoxybenzyl; 3,4-dimethoxybenzyl; and nitrobenzyl); carbonyl-protecting groups (e.g., acetal and ketal groups, such as dimethyl acetal, 1,3 -di oxolane, and the like; acylal groups; and dithiane groups, such as 1,3-dithianes, 1,3-dithiolane, and the like); carboxylic acid-protecting groups (e.g., ester groups, such as methyl ester, benzyl ester, t-butyl ester, orthoesters, and the like; and oxazoline groups.

The term “oxo” as used herein, represents =0.

The term “polyethylene glycol,” as used herein, represents an alkoxy chain comprised of one or more monomer units, each monomer unit consisting of-OC hC h-. Polyethyelene glycol (PEG) is also sometimes referred to as polyethylene oxide (PEO) or polyoxyethylene (POE), and these terms may be considered interchangeable for the purpose of this disclosure. For example, a polyethylene glycol may have the structure, -(CH2) S 2(OCH2CH2)si(CH2) S 3O-, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), and each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10). Polyethylene glycol may also be considered to include an aminopolyethylene glycol of -NR N1 (CH2)S2(CH 2 CH 2 O)SI(CH2)S3NR N1 -, wherein si is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each R N1 is, independently, hydrogen or optionally substituted Ci-6 alkyl.

The term “stereoisomer,” as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric forms, all diastereomers, enantiomers and/or conformers of the basic molecular structure. Some compounds may exist in different tautomeric forms, all of the latter being included within the scope of the present disclosure.

The term “sulfonyl,” as used herein, represents an -S(O)2- group.

The term “thiol,” as used herein represents an -SH group.

Biological terms

ATM stands for ataxia-telangiectasia mutated

ATR stands for ataxia telangiectasia and Rad3-related

BRCA stands for breast cancer gene

Chkl stands for checkpoint kinase 1

Chk2 stands for checkpoint kinase 2

DDR stands for DNA damage response

DNA stands for deoxyribonucleic acid DNA-PK stands for DNA-dependent protein kinase

NTSR1 stands for neurotensin receptor 1

PARP stands for poly-ADP ribose polymerase

PTEN stands for phosphatase and tensin homolog deleted on chromosome 10

WEE1 represents WEE1 G2 checkpoint kinase

Other terms

As used herein, the term “about” or “approximately” refers to a ±10% variation from the recited quantitative value (and includes the recited quantitative value itself) unless otherwise indicated or inferred from the context. For example, unless otherwise stated or inferred from the context, a dose of about 100 kBq/kg indicates a dose range of 100±10% kBq/kg, i.e., from 90 kBq/kg to 110 kBq/kg, inclusive.

As used herein, the term “administered in combination,” “combined administration,” or “co-administered” means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. Thus, two or more agents that are administered in combination need not be administered together. In some embodiments, they are administered within 90 days (e.g., within 80, 70, 60, 50, 40, 30, 20, 10, 5, 4, 3, 2, or 1 day(s)), within 28 days (e.g., with 14, 7, 6, 5, 4, 3, 2, or 1 day(s), within 24 hours (e.g., 12, 6, 5, 4, 3, 2, or 1 hour(s), or within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently closely together such that a combinatorial effect is achieved.

As used herein, “administering” an agent to a subject includes contacting cells of said subject with the agent.

The term “cancer” refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas. A “solid tumor cancer” is a cancer comprising an abnormal mass of tissue, e.g., sarcomas, carcinomas, and lymphomas. A “hematological cancer” or “liquid cancer,” as used interchangeably herein, is a cancer present in a body fluid, e.g., lymphomas and leukemias.

The term “chelate” as used herein, refers to an organic compound or portion thereof that can be bonded to a central metal or radiometal atom at two or more points.

The term “conjugate,” as used herein, refers to a molecule that contains a chelating group or metal complex thereof, a linker group, and which optionally contains a therapeutic moiety or a targeting moiety. As used herein, the term “compound,” is meant to include all stereoisomers, geometric isomers, and tautomers of the structures depicted.

The compounds described herein can be asymmetric (e.g, having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual sub-combination of the members of such groups and ranges. For example, the term “Ci-6 alkyl” is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and Ce alkyl. Herein a phrase of the form “optionally substituted X” (e.g, optionally substituted alkyl) is intended to be equivalent to “X, wherein X is optionally substituted” (e.g, “alkyl, wherein said alkyl is optionally substituted”). It is not intended to mean that the feature “X” (e.g, alkyl) per se is optional.

As used herein, the terms “decrease,” “decreased,” “increase,” “increased,” or “reduction,” “reduced,” (e.g., in reference to therapeutic outcomes or effects) have meanings relative to a reference level. In some embodiments, the reference level is a level as determined by the use of said method with a control in an experimental animal model or clinical trial. In some embodiments, the reference level is a level in the same subject before or at the beginning of treatment. In some embodiments, the reference level is the average level in a population not being treated by said method of treatment.

The term an “effective amount” of an agent (e.g., any of the foregoing conjugates), as used herein, is that amount sufficient to effect beneficial or desired results, such as clinical results, and, as such, an “effective amount” depends upon the context in which it is being applied.

The term “lower effective dose,” when used as a term in conjunction with an agent (e.g., a therapeutic agent) refers to a dosage of the agent which is effective therapeutically in the combination therapies of the invention and which is lower than the dose which has been determined to be effective therapeutically when the agent is used as a monotherapy in reference experiments or by virtue of other therapeutic guidance.

The term “pharmaceutical composition,” as used herein, represents a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition is manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other formulation described herein.

A “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being nontoxic and noninflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, radioprotectants, sorbents, suspending or dispersing agents, sweeteners, or waters of hydration. Exemplary excipients include, but are not limited to: ascorbic acid, histidine, phosphate buffer, butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (com), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

The term “pharmaceutically acceptable salt,” as use herein, represents those salts of the compounds described here that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, or allergic response. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and m ' Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting the free base group with a suitable organic acid.

Compounds may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of compounds, be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well- known in the art, such as hydrochloric, sulphuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, among others. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethyl ammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.

The term “radiopharmaceutical” or “radioconjugate,” as used herein, refers to any compound or conjugate that includes a radioisotope or radionuclide, such as any of the radioisotopes or radionuclides described herein.

As used herein, the term “radionuclide,” refers to an atom capable of undergoing radioactive decay (e.g., 3 H, 14 C, 15 N, 18 F, 35 S, 47 Sc, 55 Co, 60 Cu, 61 Cu, 62 Cu, 64 Cu, 67 Cu, 75 Br, 76 Br , 77 Br , 89 Zr, 86 Y, 87 Y, 90 Y, 97 RU, 99 TC, " m Tc 105 Rh, 109 Pd, m In, 123 1, 124 1, 125 1, 131 1, 149 Pm, i49 Tb , 153 Sm, 166 Ho, 177 Lu, 186 Re, 188 Re, 198 Au, 199 Au, 203 Pb, 211 At, 212 Pb , 212 Bi, 213 Bi, 223 Ra, 225 Ac, 227 Th, 229Th , 66 Ga, 67 Ga, 68 Ga, 82 Rb, 117m Sn, 2O1 T1). The terms radioactive nuclide, radioisotope, or radioactive isotope may also be used to describe a radionuclide. Radionuclides may be used as detection agents. In some embodiments, the radionuclide is an alpha-emitting radionuclide. Exemplary radionuclides used in the method of this invention include, but are not limited to, 64 Cu, 67 Cu, 68 Ga, 90 Y, 149 Tb, 153 Sm, 177 Lu, 211 At, 212 Bi, 212 Pb, 213 Bi, 223 Ra, 225 Ac, and 227 Th.

As used herein, and as well understood in the art, “to treat” a condition or “treatment” of the condition (e.g., the conditions described herein such as cancer) is an approach for obtaining beneficial or desired results, such as clinical results. Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable. In the context of cancer treatment, “ameliorating” may include, for example, reducing incidence of metastases, reducing tumor volume, reducing tumor vascularization and/or reducing the rate of tumor growth. “Palliating” a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.

Chelators

The compounds of Formula I comprise chelating moieties or chelators. Exemplary chelators include, but are not limited to, DOTA, DOTAGA, NOTA, DTP A, TETA, EDTA, NOD AGA, NODASA, TRITA, CDTA, BAT, DFO and HYNIC, which are defined as below: DOTA stands for l,4,7,10-tetrazacyclododecane-l,4,7,10-tetraacetic acid, DOTAGA stands for 1,4, 7, 10-tetraazacyclododececane,l -(glutaric acid)-4,7,10- triacetic acid,

NOTA stands for 1,4,7-triazacyclononanetriacetic acid,

DTPA stands for diethylenetriaminepentaacetic acid,

TETA stands for l,4,8,l l-tetraazacyclododecane-l,4,8,l l-tetraacetic acid,

EDTA stands for ethylenediamine-N,N' -tetraacetic acid,

NODAGA stands for 1,4,7-triazacyclononane-N-glutaric acid-N',N" -diacetic acid, NODASA stands for 1,4,7- triazacyclononane -1 -succinic acid-4, 7-diacetic acid, TRITA stands for 1,4,7,10 tetraazacyclotridecane-l,4,7,10-tetraacetic acid, CDTA stands for /ra -l .2-diaminocyclohexane-N.N.N'.N'-tetraacetic acid, DFO stands for the Desferal or Desferrioxamine type group of chelators, the chemical name of the non-limiting example is N-[5-({3-[5-(Acetyl-hydroxy-amino)-pentylcarbamoyl]- propionyl}-hydroxy-amino)-pentyl]-N'-(5-amino-pentyl)-N'-hyd roxy-succinamide,

BAT stands for the Bisamino-bisthiol group of chelators, the chemical name of the non limiting example is l-[2-(2-mercapto-2-methyl-propylamino)-ethylamino]-2-methyl- propane- 2-thiol,

HYNIC stands for 6-Hydrazino-nicotinic acid, and with the chemical structures thereof being as follows:

Radiopharmaceuticals

Radiopharmaceuticals suitable for use in accordance with the present disclosure generally comprise a radionuclide chelated with a compound of Formula I, each variable as defined in the SUMMARY section above:

Chelating moieties Examples of suitable chelating moieties include, but are not limited to, DOTA (l,4,7,10-tetrazacyclododecane-l,4,7,10-tetraacetic acid), DOTAGA (1,4,7,10- tetraazacyclododececane,! -(glutaric acid)-4,7,10-triacetic acid, NOTA (1,4,7- triazacyclononanetriacetic acid), DTPA (diethylenetriaminepentaacetic acid), TETA (l,4,8,l l-tetraazacyclododecane-l,4,8,l l-tetraacetic acid), EDTA (ethylenediamine-N,N'- tetraacetic acid), NOD AGA (1,4,7-triazacyclononane-N-glutaric acid-N',N"-diacetic acid), NODASA (1,4,7- triazacyclononane -1-succinic acid-4, 7-diacetic acid), TRITA (1,4,7,10 tetraazacyclotridecane-l,4,7,10-tetraacetic acid), CDTA (fra«s-l,2-diaminocyclohexane- N,N,N',N'-tetraacetic acid), DFO (the Desferal or Desferrioxamine type group of chelators, the chemical name of the non-limiting example is N-[5-({3-[5-(Acetyl-hydroxy-amino)- pentylcarbamoyl]-propionyl}-hydroxy-amino)-pentyl]-N'-(5-ami no-pentyl)-N' -hydroxysuccinamide), BAT (the Bisamino-bisthiol group of chelators, the chemical name of the nonlimiting example is l-[2-(2-mercapto-2-methyl-propylamino)-ethylamino]-2-methyl- propane- 2-thiol), and HYNIC (6-Hydrazino-nicotinic acid).

In some embodiments, the chelating moiety is DOTA (1,4,7,10-tetraazacyclododecane- 1,4,7,10-tetraacetic acid).

Radionuclides

The present disclosure includes use of radiopharmaceuticals each comprising a radionuclide. Examples of suitable radionuclides include, but are not limited to, 47 Sc, 55 Co, 60 Cu, 61 Cu, 62 Cu, 64 Cu, 66 Ga, 67 Ga, 67 Cu, 68 Ga, 69 Er, 77 As, 82 Rb, 89 Zr, 86 Y, 87 Y, 90 Y, 97 Ru, "Tc, " m Tc, 105 Rh, 109 Pd, m In, m Ag, 121 Sn, 127 Te, 142 Pr, 143 Pr, 149 Pm, 149 Tb, 151 Pm, 159 Gd, 153 Sm, 161 Tb, 166 Dy, 166 HO, 169 Yb, 172 Tm, 175 Yb, 177 Lu, 117m Sn, 177m Sn, 186 Re, 188 Re, 188 Rd, 198 Au, 199 Au, 2O1 T1, 203 Pb, 211 At, 212 Pb , 212 Bi, 213 Bi, 223 Ra, 225 Ac, 227 Th, and 229 Th.

In some embodiments, the radionuclide is selected from the group consisting of 64 Cu, 67 Cu, 68 Ga, 90 Y, 149 Tb, 153 Sm, 177 Lu, 211 At, 212 Bi, 212 Pb, 213 Bi, 223 Ra, 225 Ac, and 227 Th.

In some embodiments, the radionuclide is an alpha emitter, e.g., Astatine-211 ( 211 At), Bismuth-212 ( 212 Bi), Bismuth-213 ( 213 Bi), Actinium-225 ( 225 Ac), Radium-223 ( 223 Ra), Lead- 212 ( 212 Pb), Thorium-227 ( 227 Th), or Terbium-149 ( 149 Tb). Linkers

The radiopharmaceuticals used in the methods of the present disclosure comprise the linker as shown within the structure of Formula I that comprises Li and L2: wherein Li is C2-5 alkylidene; L2 is C2-20 alkylidene, C2-20 heteroalky li dene, (C=O)O, (C=O)NR, or a combination thereof, R being hydrogen or C1-4 alkyl.

An exemplary optionally substituted with a substituent described herein.

L2 typically comprises at least one heteroatom (e.g., O or N), amide moiety, or both. Exemplary L2 includes, but is not limited to, the following: optionally substituted with a substituent described herein.

DNA Damage and Repair Inhibitors (DDRi)

As disclosed herein, the terms “DNA Damage Response inhibitor” and “DNA Damage and Repair inhibitor” are used interchangeably. In various embodiments, a DNA Damage and Repair inhibitor (DDRi) is co-administered with a radiopharmaceutical.

DNA repair involves multiple molecular pathways that repair DNA single strand breaks (e.g., the PARP pathway) and double stranded breaks (e.g., BRCA and other genes such as ATR/ATM). PARP inhibition (PARPi) results in a failure of single stranded break repair, which further leads to double stranded breaks. Available PARP inhibitors act through both PARP enzyme inhibition and DNA-trapping. Tumor cells with BRCA and/or PTEN mutations are sensitive to PARPi. ATR inhibition (ATRi) results in a failure to repair double stranded breaks — the accumulation of double stranded breaks results in cell death. These inhibitors act by preventing homologous recombination and non-homologous end joining mechanisms.

The present disclosure relates to combination therapy with radiopharmaceuticals and DNA Damage and Repair inhibitors. It has been found that this type of combination therapy results in unexpected improvement in the treatment of cancer, especially in cancers that would not be expected to be responsive to the DDRi.

In some embodiments, the DDRi is a PARP inhibitor (PARPi). In some embodiments, the PARPi is selected from the group consisting of niparib, niraparib, olaparib, pamiparib, rucaparib (camsylate), talazoparib, and veliparib, or an analog thereof. In some embodiments, the PARPi is adavosertib, AZD2811, or an analog thereof.

In some embodiments, the DDRi is an ATM/ATR inhibitor. In some embodiments the ATM/ATR inhibitor is selected from the group consisting of KLI , AZD0156, AZDI 390, AZD6738, BAY-1895344, EPT-46464, M3541, M4344, M6620 (formerly known as VE-922 or VX-970), NU6027, and VE-821, or an analog thereof. In certain embodiments, the ATM/ATR inhibitor is AZD1390 or an analog thereof.

In some embodiments, the DDRi is a WEE1 inhibitor, a Chkl inhibitor, or a Chk2 inhibitor. Examples of a WEE1 inhibitor, a Chkl inhibitor, or a Chk2 inhibitor include those known in the field.

In some embodiments, the DDRi is a DNA-dependent protein kinase (DNA-PK) inhibitor. Non-limiting examples of DNA-PK inhibitors include, but are not limited to, AZD7648, KU-0060648, NU7026, NU7441 (KU-57788), PI-103, PIK-75 HCI, PP121, SF2523, and analogs thereof. In certain embodiments, the DNA-PK inhibitor is AZD7648 or an analog thereof.

Subjects

In some disclosed methods, a therapy (e.g., comprising a therapeutic agent) is administered to a subject. In some embodiments, the subject is a mammal, e.g., a human.

In some embodiments, the subject has received or is receiving another therapy. For example, in some embodiments, the subject has received or is receiving a radiopharmaceutical. In some embodiments, the subject has received or is receiving a DDRi.

In some embodiments, the subject has cancer or is at risk of developing cancer. For example, the subject may have been diagnosed with cancer. The cancer may be a primary cancer or a metastatic cancer. Subjects may have any stage of cancer, e.g., stage I, stage II, stage III, or stage IV with or without lymph node involvement and with or without metastases. Provided compositions may prevent or reduce further growth of the cancer and/or otherwise ameliorate the cancer (e.g., prevent or reduce metastases). In some embodiments, the subject does not have cancer but has been determined to be at risk of developing cancer, e.g., because of the presence of one or more risk factors such as environmental exposure, presence of one or more genetic mutations or variants, family history, etc. In some embodiments, the subject has not been diagnosed with cancer.

In some embodiments, the cancer is a solid tumor.

In some embodiments, the solid tumor cancer is breast cancer, non-small cell lung cancer, small cell lung cancer, pancreatic cancer, head and neck cancer, prostate cancer, colorectal cancer, sarcoma, adrenocortical carcinoma, neuroendocrine cancer, Ewing's Sarcoma, multiple myeloma, or acute myeloid leukemia.

In some embodiments, the cancer is a non-solid (e.g., liquid (e.g., hematologic)) cancer.

Administration and dosage

Effective and lower effective doses

The present disclosure provides combination therapies in which the amounts of each therapeutic may or may not be, on their own, therapeutically effective. For example, provided are methods comprising administering a first therapy and a second therapy in amounts that together are effective to treat or ameliorate a disorder, e.g., cancer. In some embodiments, at least one of the first and second therapies is administered to the subject in a lower effective dose. In some embodiments, both the first and the second therapies are administered in lower effective doses.

In some embodiments, the first therapy comprises a radiopharmaceutical and the second therapy comprises a DDRi.

In some embodiments, the first therapy comprises a DDRi and the second therapy comprises a radiopharmaceutical.

In some embodiments, therapeutic combinations as disclosed herein are administered to a subject in a manner (e.g., dosing amount and timing) sufficient to cure or at least partially arrest the symptoms of the disorder and its complications. In the context of a single therapy (a “monotherapy”), an amount adequate to accomplish this purpose is defined as a “therapeutically effective amount,” an amount of a compound sufficient to substantially improve at least one symptom associated with the disease or a medical condition. The “therapeutically effective amount” typically varies depending on the therapeutic. For known therapeutic agents, the relevant therapeutically effective amounts may be known to or readily determined by those of skill in the art.

For example, in the treatment of cancer, an agent or compound that decreases, prevents, delays, suppresses, or arrests any symptom of the disease or condition would be therapeutically effective. A therapeutically effective amount of an agent or compound is not required to cure a disease or condition but will provide a treatment for a disease or condition such that the onset of the disease or condition is delayed, hindered, or prevented, or the disease or condition symptoms are ameliorated, or the term of the disease or condition is changed or, for example, is less severe or recovery is accelerated in an individual. For example, a treatment may be therapeutically effective if it causes a cancer to regress or to slow the cancer’s growth.

The dosage regimen (e.g., amounts of each therapeutic, relative timing of therapies, etc.) that is effective for these uses may depend on the severity of the disease or condition and the weight and general state of the subject. For example, the therapeutically effective amount of a particular composition comprising a therapeutic agent applied to mammals (e.g., humans) can be determined by the person of ordinary skill in the art with consideration of individual differences in age, weight, and the condition of the mammal. Because certain conjugates of the present disclosure exhibit an enhanced ability to target cancer cells and residualize, the dosage of these compounds can be lower than (e.g., less than or equal to about 90%, 75%, 50%, 40%, 30%, 20%, 15%, 12%, 10%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of) the equivalent dose of required for a therapeutic effect of the unconjugated agent. Therapeutically effective and/or optimal amounts can also be determined empirically by those of skill in the art. Thus, lower effective doses can also be determined by those of skill in the art.

Single or multiple administrations of a radiopharmaceutical or a composition (e.g., a pharmaceutical composition comprising a therapeutic agent or a radiopharmaceutical) can be carried out with dose levels and pattern being selected by the treating physician. The dose and administration schedule can be determined and adjusted based on the severity of the disease or condition in the subject, which may be monitored throughout the course of treatment according to the methods commonly practiced by clinicians or those described herein.

In the disclosed combination therapy methods, the first and second therapies may be administered sequentially or concurrently to a subject. For example, a first composition comprising a first therapeutic agent and a second composition comprising a second therapeutic agent may be administered sequentially or concurrently to a subject. Alternatively, a composition comprising a combination of a first therapeutic agent and a second therapeutic agent may be administered to the subject.

In some embodiments, the radiopharmaceutical is administered in a single dose. In some embodiments, the radiopharmaceutical is administered more than once, i.e., multiple doses. When the radiopharmaceutical is administered more than once, the dose of each administration may be the same or different.

In some embodiments, the DDRi is administered in a single dose. In some embodiments, the DDRi is administered more than once, e.g., at least twice, at least three times, etc. In some embodiments, the DDRi is administered multiple times according to a regular or semi-regular schedule, e.g., once every approximately two weeks, once a week, twice a week, three times a week, or more than three times a week. When the DDRi is administered more than once, the dose of each administration may be the same or different. For example, the DDRi may be administered in an initial dose amount, and then subsequent dosages of the DDRi may be higher or lower than the initial dose amount.

In some embodiments, the first dose of the DDRi is administered at the same time as the first dose of the radiopharmaceutical. In some embodiments, the first dose of the DDRi is administered before the first dose of radiopharmaceutical. In some embodiments, the first dose of the DDRi is administered after the first dose of radiopharmaceutical. In some embodiments, subsequent doses of the DDRi are administered.

In some embodiments, the present disclosure provides methods comprising administering to a mammal an 225 Ac-radiopharmaceutical at a dosage of less than 1 MBq/kg (e.g., less than 800 kBq/kg, less than 600 kBq/kg, less than 500 kBq/kg, less than 400 kBq/kg, less than 300 kBq/kg, less than 250 kBq/kg, less than 200 kBq/kg, less than 150 kBq/kg, less than 100 kBq/kg, or less than 50 kBq/kg) of body weight of said mammal. Each of the dose may be administered multiple times to the mammal.

In certain embodiments, said 225 Ac-radiopharmaceutical can be administered at a dosage of between 900 kBq/kg and 800 kBq/kg, between 800 kBq/kg and 700 kBq/kg, between 700 kBq/kg and 600 kBq/kg, between 600 kBq/kg and 500 kBq/kg, between 500 kBq/kg and 400 kBq/kg, between 400 kBq/kg and 300 kBq/kg, between 300 kBq/kg and 200 kBq/kg, between 200 kBq/kg and 100 kBq/kg, or between 100 kBq/kg and 50 kBq/kg. Each of the dose may be administered multiple times to the mammal.

In certain embodiments, said 225 Ac-radiopharmaceutical can be administered at a dosage of about 2 MBq/kg, about 1.9 MBq/kg, about 1.8 MBq/kg, about 1.7 MBq/kg, about 1.6 MBq/kg, about 1.5 MBq/kg, about 1.4 MBq/kg, about 1.3 MBq/kg, about 1.2 MBq/kg, about 1.1 MBq/kg, about 1 MBq/kg, about 0.9 MBq/kg, about 0.8 MBq/kg, about 0.7 MBq/kg, about 0.6 MBq/kg, about 0.5 MBq/kg, about 0.4 MBq/kg, about 0.3 MBq/kg, about 0.2 MBq/kg, about 0.1 MBq/kg, or about 0.05 MBq/kg. Each of the dose may be administered multiple times to the mammal. In some embodiments, said 225 Ac-radiopharmaceutical is administered at a dosage of less than 250 kBq/kg (e.g., about 240 kBq/kg, about 220 kBq/kg, about 200 kBq/kg, about 180 kBq/kg, about 160 kBq/kg, about 150 kBq/kg, about 140 kBq/kg, about 130 kBq/kg, about 120 kBq/kg, about 110 kBq/kg, or about 100 kBq/kg) of body weight of said mammal. Each of the dose may be administered multiple times to the mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered at a dosage of less than 100 kBq/kg (e.g., about 90 kBq/kg, about 80 kBq/kg, about 70 kBq/kg, about 60 kBq/kg, about 50 kBq/kg, about 40 kBq/kg, about 30 kBq/kg, about 20 kBq/kg, or about 10 kBq/kg) of body weight of said mammal. Each of the dose may be administered multiple times to the mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 15 MBq (e.g., about 14 MBq, about 13 MBq, about 12 MBq, about 11 MBq, about 10 MBq, about 9 MBq, about 8 MBq, about 7 MBq, about 6 MBq, about 5 MBq, about 4 MBq, about 3 MBq, about 2 MBq, about 1 MBq) to said mammal. Each of the unitary dosage may be administered multiple times to the mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 10 MBq to said mammal. Each of the unitary dosage may be administered multiple times to the mammal.

In some embodiments, said 225 Ac-radiopharmaceutical is administered as a unitary dosage of less than 5 MBq to said mammal. Each of the unitary dosage may be administered multiple times to the mammal.

In some embodiments, radiopharmaceuticals (or a composition thereof) and DDRis (or a composition thereof) are administered within 28 days (e.g., within 14, 7, 6, 5, 4, 3, 2, or 1 day(s)) of each other.

In some embodiments, radiopharmaceuticals (or a composition thereof) and DDRis (or a composition thereof) are administered within 90 days (e.g., within 80, 70, 60, 50, 40, 30, 20, 10, 5, 4, 3, 2, or 1 day(s)) of each other. In various embodiments the DDRi is administered at the same time as radiopharmaceutical. In various embodiments, the DDRi is administered multiple times after the first administration of radiopharmaceutical.

In some embodiments, compositions (such as compositions comprising radiopharmaceuticals) are administered for radiation treatment planning or diagnostic purposes. When administered for radiation treatment planning or diagnostic purposes, compositions may be administered to a subject in a diagnostically effective dose and/or an amount effective to determine the therapeutically effective dose. In some embodiments, a first dose of disclosed conjugate or a composition (e.g., pharmaceutical composition) thereof is administered in an amount effective for radiation treatment planning, followed administration of a combination therapy including a conjugate as disclosed herein and another therapeutic.

Pharmaceutical compositions comprising one or more agents (e.g., radiopharmaceuticals and/or DDRis) can be formulated for use in accordance with disclosed methods and systems in a variety of drug delivery systems. One or more physiologically acceptable excipients or carriers can also be included in the composition for proper formulation. Examples of suitable formulations are found in Remington ’s Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 17th ed., 1985. For a brief review of methods for drug delivery, see, e.g., Langer (Science 249:1527-1533, 1990).

Formulations

Pharmaceutical compositions may be formulated for parenteral, intranasal, topical, oral, or local administration, such as by atransdermal means, for prophylactic and/or therapeutic treatment. Pharmaceutical compositions can be administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion, or by topical application or intraarticular injection at areas affected by the vascular or cancer condition. Examples of additional routes of administration include intravascular, intra-arterial, intratumor, intraperitoneal, intraventricular, intraepidural, as well as nasal, ophthalmic, intrascleral, intraorbital, rectal, topical, or aerosol inhalation administration. Also specifically contemplated are sustained release administration, by such means as depot injections or erodible implants or components. Suitable compositions include compositions comprising include agents (e.g., compounds as disclosed herein) dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, or PBS, among others, e.g., for parenteral administration. Compositions may contain pharmaceutically acceptable auxiliary substances to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, or detergents, among others. In some embodiments, compositions are formulated for oral delivery; for example, compositions may contain inert ingredients such as binders or fillers for the formulation of a unit dosage form, such as a tablet or a capsule. In some embodiments, compositions are formulated for local administration; for example, compositions may contain inert ingredients such as solvents or emulsifiers for the formulation of a cream, an ointment, a gel, a paste, or an eye drop. Compositions may be sterilized, e.g., by conventional sterilization techniques, or sterile filtered. Aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 6 and 7, such as 6 to 6.5. In some embodiments, compositions in solid form are packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules. In some embodiments, compositions in solid form are packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.

Effects

In some embodiments, methods of the present disclosure result in a therapeutic effect. In some embodiments, the therapeutic effect comprises a decrease in tumor volume, a stable tumor volume, or a reduced rate of increase in tumor volume. In some embodiments, the therapeutic effect comprises a decreased incidence of recurrence or metastasis.

Other agents

In some embodiments, disclosed methods further include administering an antiproliferative agent, radiation sensitizer, or an immunoregulatory or immunomodulatory agent.

By “antiproliferative” or “antiproliferative agent,” as used interchangeably herein, is meant any anticancer agent, including those antiproliferative agents listed in Table 1, any of which can be used in combination with a radiopharmaceutical to treat a condition or disorder. Antiproliferative agents also include organo-platinum derivatives, naphtoquinone and benzoquinone derivatives, chrysophanic acid and anthraquinone derivatives thereof.

By “immunoregulatory agent” or “immunomodulatory agent,” as used interchangeably herein, is meant any immuno-modulator, including those listed in Table 1, any of which can be used in combination with a radiopharmaceutical provided herein.

As used herein, “radiation sensitizer” includes any agent that increases the sensitivity of cancer cells to radiation therapy. Radiation sensitizers may include, but are not limited to, 5- fluorouracil, analogs of platinum (e.g., cisplatin, carboplatin, oxaliplatin), gemcitabine, EGFR antagonists (e.g., cetuximab, gefitinib), famesyltransferase inhibitors, COX-2 inhibitors, bFGF antagonists, and VEGF antagonists.

Examples

Example 1. Synthesis of Radiopharmaceuticals Comprising Compounds of Formula I Compounds of Formula I are small molecule antagonists targeting NTSR1, which can be radiolabeled with a radionuclide such as Lutetium-177 ( 177 Lu) or Actinium-225 ( 225 Ac) to form radionuclide-chelated radiopharmaceuticals. The synthesis of compounds of Formula I, or their radionuclide-chelated radiopharmaceuticals, can be referred to US Patent No. 10,961,199 B2.

The following exemplary compound, i.e., Compound A, prepared according to US Patent No. 10,961,199 B2 was used in the in vivo studies provided in Examples 2-5 below.

Example 2. [ 177 Lu] -Compound A Biodistribution in the CT-26-mNTSRl Syngeneic Immunocompetent Mouse Model

Compound A of Formula I was radiolabeled with Lu-177 using methods well known in the art to form [ 177 Lu] -Compound A. The ability of [ 177 Lu] -Compound A to target antigen expressing mouse NTSR1 overexpressing tumors in vivo was demonstrated using the CT-26 syngeneic model. Tumor uptake was maintained at 7-2.85 % injected dose/g (ID/g) from 6- 48 hours post injection. See Figure 1.

Example 3. Single Agent Efficacy of [ 225 Ac]-Compound A in CT-26-mNTSRl Xenograft Model

Compound A of Formula I was radiolabeled using standard techniques to form [ 225 Ac] -Compound A. An efficacy study of [ 225 Ac] -Compound A in immunocompetent mice was conducted using various doses of [ 225 Ac] -Compound A, ranging from 0.185 to 5.555 MBq/kg (single-dose, intravenous). It was found that [ 225 Ac] -Compound A had enhanced efficacy in reducing tumor volume in CT-26-mNTSRl xenograft bearing mice relative to the efficacy of cold Compound A. See Figure 2. Example 4. Combination of [ 225 Ac] -Compound A and Olaparib treatment in CT-26- mNTSRl xenograft model Results in Increased Therapeutic Efficacy

An in vivo study was conducted to test the effect of [ 225 Ac] -Compound A (as described in Example 3) in combination with Olaparib in CT-26-mNTSRl tumor bearing mice. Mice treated with either [ 225 Ac] -Compound A at 0.555 MBq/kg or olaparib at 50 mg/kg showed slight or neglible reduction in tumor growth compared to control mice. However, when [ 225 Ac] -Compound A was co-administered at a 0.555 MBq/kg dose (single dose, intravenous) with Olaparib (25mg/kg QD; oral), increased therapeutic efficacy, including tumor regression, was observed compared to vehicle only or monotherapy ([ 225 Ac]- Compound A only or olaparib only) treatment groups. See Figure 3.

Example 5. Improvement of the Overall Survival in [ 225 Ac] -Compound A treated Mice

An in vivo study was conducted to test the effect of [ 225 Ac] -Compound A (as described in Example 3) and olaparib, on survival in the CT-26-mNTSRl mouse model. When [ 225 Ac] -Compound A was co-administered at a 0.555 MBq/kg dose (single-dose intravenous) with olaparib (25 mg/kb, QD, oral), improved overall survival was observed — co-administration resulted in significantly improved survival when compared to vehicle control, the [ 225 Ac] -Compound A-treated, or olaparib-treated groups. See Figure 4.

OTHER EMBODIMENTS

Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments described herein. Such equivalents are intended to be encompassed by the following claims.