WO2017205447A1 - Méthodes de traitement du cancer avec un composé ligand de psma-tubulysine - Google Patents

Méthodes de traitement du cancer avec un composé ligand de psma-tubulysine Download PDF

Info

Publication number
WO2017205447A1
WO2017205447A1 PCT/US2017/034130 US2017034130W WO2017205447A1 WO 2017205447 A1 WO2017205447 A1 WO 2017205447A1 US 2017034130 W US2017034130 W US 2017034130W WO 2017205447 A1 WO2017205447 A1 WO 2017205447A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
therapeutically effective
effective amount
imaging
aspects
Prior art date
Application number
PCT/US2017/034130
Other languages
English (en)
Inventor
Christopher Paul Leamon
Binh Nguyen
Original Assignee
Endocyte, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Endocyte, Inc. filed Critical Endocyte, Inc.
Publication of WO2017205447A1 publication Critical patent/WO2017205447A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention described herein pertains to drug delivery conjugates for targeted therapy.
  • the invention described herein relates to methods of treating PSMA expressing cancers with a PSMA ligand-tubulysin compound.
  • the invention described herein also relates to methods of treating PSMA-expressing cancers with a PSMA ligand-tubulysin compound in patients where stable disease results after treatment with the PSMA ligand-tubulysin compound.
  • Prostate specific membrane antigen is a type II cell surface membrane-bound glycoprotein with -110 kD molecular weight, including an intracellular segment (amino acids 1-18), a transmembrane domain (amino acids 19-43), and an extensive extracellular domain (amino acids 44-750). While the functions of the intracellular segment and the transmembrane domains are currently believed to be insignificant, the extracellular domain is involved in several distinct activities. PSMA plays a role in the central nervous system, where it metabolizes N-acetyl-aspartyl glutamate (NAAG) into glutamic and N-acetyl aspartic acid.
  • NAAG N-acetyl-aspartyl glutamate
  • PSMA is also sometimes referred to as an N-acetyl alpha linked acidic dipeptidase (NAALADase).
  • PSMA is also sometimes referred to as a folate hydrolase I (FOLH I) or glutamate carboxypeptidase (GCP II) due to its role in the proximal small intestine where it removes ⁇ -linked glutamate from poly-y-glutamated folate and a-linked glutamate from peptides and small molecules.
  • FOLH I folate hydrolase I
  • GCP II glutamate carboxypeptidase
  • PSMA is named largely due to its higher level of expression on prostate cancer cells; however, its particular function on prostate cancer cells remains unresolved.
  • PSMA is over- expressed in the malignant prostate tissues when compared to other organs in the human body such as kidney, proximal small intestine, and salivary glands. Unlike many other membrane- bound proteins, PSMA undergoes rapid internalization into the cell in a similar fashion to cell surface bound receptors like vitamin receptors. PSMA is internalized through clathrin-coated pits and subsequently can either recycle to the cell surface or go to lysosomes. It has been suggested that the dimer and monomer form of PSMA are inter-convertible, though direct evidence of the interconversion is being debated. Even so, only the dimer of PSMA possesses enzymatic activity, and the monomer does not.
  • PSMA represents a viable target for the selective and/or specific delivery of biologically active agents, including drug compounds and imaging agents to such prostate cells.
  • biologically active agents including drug compounds and imaging agents to such prostate cells.
  • One such drug compound is tubulysin, which when conjugated to PSMA through an appropriately functionalized linker provides
  • Imaging agent is the PSMA ligand-imaging conjugate of formula Ilia
  • Imaging conjugate Ilia has found use as a cancer imaging agent as described in, for example, WO2009/026177.
  • the present disclosure provides a method for treating a cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the use comprises administering to the patient a therapeutically effective amount of the PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the medicament comprises a therapeutically effective amount of the PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof.
  • the cancer is a PSMA expressing cancer. In some aspects of these embodiments, the compound is at least about 98 percent pure. In some embodiments, the cancer is selected from the group consisting of a glioma, a carcinoma, a sarcoma, a lymphoma, a melanoma, a mesothelioma, a nasopharyngeal carcinoma, a leukemia, an adenocarcinoma, and a myeloma.
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non- small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic leukemia, acute leukemia, lymphocytic lymphomas, pleural mesothelioma, cancer
  • the cancer is a primary or secondary brain cancer. In some aspects of these embodiments, the cancer is prostate cancer. In some aspects of these embodiments, the cancer is metastatic prostate cancer. In some aspects of these embodiments, the PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof, is administered in a parenteral dosage form. In some aspects of these embodiments, the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 100.0 mg/m 2.
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 90.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 80.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 70.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 60.0 mg/m 2. In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 40.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 35.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 25.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 24.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 24.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 23.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 23.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 22.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 22.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 21.5 mg/m 2. In some aspects of these embodiments,
  • the therapeutically effective amount is from about 0.1 mg/m to about 21.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 20.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 20.0 mg/m 2. In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 19.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 18.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 18.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 17.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 17.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 16.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 15.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 15.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 14.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 14.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 13.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 13.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 12.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2
  • the therapeutically effective amount is from about 0.1 mg/m to about 12.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 11.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 11.0 mg/m 2. In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 10.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 9.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 8.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 8.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 7.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 7.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 6.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 6.0 mg/m 2.
  • the therapeutically effective amount is from about 0.1 mg/m to about 5.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 5.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 2.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 2.0 mg/m 2 .
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal. In some aspects of these embodiments, the therapeutically effective amount is from
  • the therapeutically effective amount is from 0.1 mg/m 2 to 100.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 90.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 80.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 70.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 60.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 50.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 40.0 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 35.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 30.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 25.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 24.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 24.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 23.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 23.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 22.5
  • the therapeutically effective amount is from 0.1 mg/m 2 to 22.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 20.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 20.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1
  • the amount is from 0.1 mg/m to 19.0 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from 0.1 mg/m to 18.5 mg/m . In some aspects of these
  • the therapeutically effective amount is from 0.1 mg/m to 18.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 17.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1
  • the amount is from 0.1 mg/m to 16.5 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from 0.1 mg/m to 16.0 mg/m . In some aspects of these
  • the therapeutically effective amount is from 0.1 mg/m to 15.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 15.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1
  • the amount is from 0.1 mg/m to 14.0 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from 0.1 mg/m to 13.5 mg/m . In some aspects of these
  • the therapeutically effective amount is from 0.1 mg/m to 13.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 12.5
  • the therapeutically effective amount is from 0.1
  • the amount is from 0.1 mg/m to 11.5 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from 0.1 mg/m to 11.0 mg/m . In some aspects of these
  • the therapeutically effective amount is from 0.1 mg/m to 10.5 mg/m .
  • the therapeutically effective amount is from 0.1 mg/m to 10.0
  • the therapeutically effective amount is from 0.1
  • the amount is from 0.1 mg/m to 9.0 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from 0.1 mg/m to 8.5 mg/m . In some aspects of these
  • the therapeutically effective amount is from 0.1 mg/m to 8.0 mg/m . In some embodiments, the therapeutically effective amount is from 0.1 mg/m to 8.0 mg/m . In some embodiments, the therapeutically effective amount is from 0.1 mg/m to 8.0 mg/m . In some embodiments, the therapeutically effective amount is from 0.1 mg/m to 8.0 mg/m . In some embodiments, the therapeutically effective amount is from 0.1 mg/m to 8.0 mg/m . In some
  • the therapeutically effective amount is from 0.1 mg/m to 7.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1
  • the therapeutically effective amount is from 0.1 mg/m to 6.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 6.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 5.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 5.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 4.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 2.5
  • the therapeutically effective amount is from 0.1 mg/m 2 to 2.0 mg/m 2.
  • the methods and uses described herein further comprise imaging PSMA expression by the cancer.
  • the step of imaging occurs before the step of administering.
  • the imaging is performed by imaging wherein the imaging is selected from the group consisting of SPECT imaging, PET imaging, IHC, and FISH.
  • the imaging is performed by SPECT imaging.
  • the step of imaging comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • the step of imaging comprises administering a PSMA ligand-imaging conjugate of the formula III or a pharmaceutically acceptable salt thereof, wherein R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein M is a cation of a radionuclide.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium. In some aspects of these embodiments, M in the conjugate, or a pharmaceutically acceptable salt thereof, is an isotope of technetium.
  • the PSMA ligand-imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the methods and uses described herein further comprise determining the PSMA status of the patient by imaging.
  • the imaging is SPECT imaging.
  • the PSMA status of the patient correlates with a clinical benefit to the patient.
  • the clinical benefit is selected from the group consisting of inhibition of tumor growth, stable disease, a partial response, and a complete response.
  • the clinical benefit is stable disease.
  • the PSMA positive lesions indicate functionally active PSMA.
  • the step of determining comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein the conjugate is bound to a radionuclide.
  • the step of determining comprises administering a PSMA li and-imaging conjugate of the formula III
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein M is a cation of a radionuclide.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium. In some aspects of these embodiments, M in the conjugate, or a pharmaceutically acceptable salt thereof, is an isotope of technetium. In some aspects of these embodiments, the PSMA ligand- imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the present disclosure provides a method of treating a cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the use comprises administering to the patient a therapeutically effective amount of the PSMA ligand-tubulysin compound I.
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the medicament comprises a therapeutically effective amount of the PSMA ligand- tubulysin compound I, or a pharmaceutically acceptable salt thereof.
  • the patient has been treated with at least one prior treatment.
  • the at least one prior treatment is selected from the group consisting of a chemotherapeutic agent, surgery, radiation therapy, immunotherapy, photodynamic therapy, stem cell therapy, and hyperthermia.
  • the at least one prior treatment is a systemic treatment.
  • the systemic treatment is selected from the group consisting of palifosfamide, 5-fluorouracil, capecitabine, pemetrexed, cisplatin, carboplatin, gemcitabine, paclitaxel, vinorelbine, eribulin, docetaxel, cyclophosphamide, doxorubicin, regorafinib, and combinations thereof.
  • the cancer is a PSMA expressing cancer.
  • the compound is at least about 98 percent pure.
  • the cancer is selected from the group consisting of a glioma, a carcinoma, a sarcoma, a lymphoma, a melanoma, a mesothelioma, a
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non-small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the pen
  • the cancer is a primary or secondary brain cancer.
  • the cancer is prostate cancer.
  • the cancer is metastatic prostate cancer.
  • the PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof is administered in a parenteral dosage form.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal.
  • the therapeutically effective amount is from about 0.1 mg/m to about 100.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount
  • therapeutically effective amount is from about 0.1 mg/m to about 80.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 50.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 40.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 30.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 25.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 24.0 mg/m . In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about 23.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 22.5 mg/m . In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 21.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 19.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 19.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 18.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 18.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 17.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 17.0 mg/m 2.
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 16.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 16.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 15.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 15.0 mg/m 2. In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about 14.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 14.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 13.5 mg/m 2. In some aspects
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 12.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 12.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 11.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 10.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 10.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 9.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 9.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 8.5 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 8.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 7.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 7.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 6.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 6.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 5.5 mg/m 2.
  • the therapeutically effective amount is from about 0.1 mg/m to about 5.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 2.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 2.0 mg/m 2.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal. In some aspects of these embodiments, the therapeutically effective amount is from
  • the therapeutically effective amount is from 0.1 mg/m 2 to 100.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 90.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 80.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 70.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 60.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 50.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 40.0 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 35.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 30.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 25.0
  • the therapeutically effective amount is from 0.1 mg/m 2 to 24.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 24.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 23.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 23.0 mg/m 2. In some
  • the therapeutically effective amount is from 0.1 mg/m to 22.5
  • the therapeutically effective amount is from 0.1 mg/m 2 to 22.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 20.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 20.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 19.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 19.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 18.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 18.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 17.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 17.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 16.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 16.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 15.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 15.0
  • the therapeutically effective amount is from 0.1 mg/m 2 to 14.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 14.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 13.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 13.0 mg/m 2. In some
  • the therapeutically effective amount is from 0.1 mg/m to 12.5
  • the therapeutically effective amount is from 0.1 mg/m 2 to 12.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 11.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 11.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 10.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 12.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 11.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 11.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 10.5 mg/m 2. In some
  • the therapeutically effective amount is from 0.1 mg/m to 10.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 9.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 9.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 8.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 8.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 7.5 mg/m .
  • the therapeutically effective amount is from 0.1 mg/m 2 to 7.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 6.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 6.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 5.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 5.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 4.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 2.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 2.0 mg/m 2.
  • the methods and uses described herein further comprise imaging PSMA expression by the cancer.
  • the step of imaging occurs before the step of administering.
  • the imaging is performed by imaging wherein the imaging is selected from the group consisting of SPECT imaging, PET imaging, IHC, and FISH.
  • the imaging is performed by SPECT imaging.
  • the step of imaging comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • the step of imaging comprises administering a PSMA ligand-imaging conjugate of the formula III or a pharmaceutically acceptable salt thereof, wherein R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein M is a cation of a radionuclide.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium. In some aspects of these embodiments, M in the conjugate, or a pharmaceutically acceptable salt thereof, is an isotope of technetium.
  • the PSMA ligand-imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the methods and uses described herein further comprise determining the PSMA status of the patient by imaging.
  • the imaging is SPECT imaging.
  • the PSMA status of the patient correlates with a clinical benefit to the patient.
  • the clinical benefit is selected from the group consisting of inhibition of tumor growth, stable disease, a partial response, and a complete response.
  • the clinical benefit is stable disease.
  • the step of determining comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • the step of determining comprises administering a PSMA ligand-imaging conjugate of the formula III
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is an isotope of technetium.
  • the PSMA ligand- imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the present disclosure provides a method of treating a PSMA expressing cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the use comprises administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides use of a PSMA ligand-tubulysin compound I
  • the medicament comprises a therapeutically effective amount of the PSMA ligand- tubulysin compound I, or a pharmaceutically acceptable salt thereof.
  • the patient has been treated with at least one prior treatment.
  • the at least one prior treatment is selected from the group consisting of a chemotherapeutic agent, surgery, radiation therapy, immunotherapy, photodynamic therapy, stem cell therapy, and hyperthermia.
  • the at least one prior treatment is a systemic treatment.
  • the systemic treatment is selected from the group consisting of palifosfamide, 5-fluorouracil, capecitabine, pemetrexed, cisplatin, carboplatin, gemcitabine, paclitaxel, vinorelbine, eribulin, docetaxel, cyclophosphamide, doxorubicin, regorafinib, and combinations thereof.
  • the cancer is a PSMA expressing cancer.
  • the compound is at least about 98 percent pure.
  • the cancer is selected from the group consisting of a glioma, a carcinoma, a sarcoma, a lymphoma, a melanoma, a mesothelioma, a
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non-small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the pen
  • the cancer is a primary or secondary brain cancer.
  • the cancer is prostate cancer.
  • the cancer is metastatic prostate cancer.
  • the PSMA ligand-tubulysin compound I, or a pharmaceutically acceptable salt thereof is administered in a parenteral dosage form.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal.
  • the therapeutically effective amount is from about 0.1 mg/m to about 100.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount
  • therapeutically effective amount is from about 0.1 mg/m to about 80.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 70.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 50.0 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about 40.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 30.0 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 24.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 22.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 22.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 21.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 20.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 19.5 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about 19.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 18.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 17.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 16.5 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 15.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 13.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 13.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 12.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 11.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 10.5 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about 10.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is
  • therapeutically effective amount is from about 0.1 mg/m to about 9.0 mg/m .
  • the therapeutically effective amount is from about 0.1 mg/m to about
  • the therapeutically effective amount is from
  • effective amount is from about 0.1 mg/m to about 7.5 mg/m . In some aspects of these
  • the therapeutically effective amount is from about 0.1 mg/m to about 7.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1
  • the amount is from about 0.1 mg/m to about 6.0 mg/m . In some aspects of these embodiments, the
  • therapeutically effective amount is from about 0.1 mg/m to about 5.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m to about 5.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 2.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from about 0.1 mg/m 2 to about 2.0 mg/m 2.
  • the parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal. In some aspects of these embodiments, the therapeutically effective amount is from
  • the therapeutically effective amount is from 0.1 mg/m 2 to 100.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 90.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 80.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 70.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 60.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 50.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 40.0 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 35.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 30.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 25.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 24.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 24.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 23.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 23.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 22.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 22.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 21.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 20.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m to 20.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 19.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 19.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 18.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 18.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 17.5 mg/m .
  • the therapeutically effective amount is from 0.1 mg/m 2 to 17.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 16.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 16.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 15.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 15.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 14.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 14.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 13.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 13.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 12.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 12.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 11.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 11.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 10.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 10.0
  • the therapeutically effective amount is from 0.1 mg/m 2 to 9.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 9.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 8.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 8.0 mg/m 2. In some
  • the therapeutically effective amount is from 0.1 mg/m to 7.5
  • the therapeutically effective amount is from 0.1 mg/m 2 to 7.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 6.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 6.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 5.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 5.0 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 4.5 mg/m 2.
  • the therapeutically effective amount is from 0.1 mg/m 2 to 4.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.5 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 3.0 mg/m 2. In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m to 2.5 mg/m . In some aspects of these embodiments, the therapeutically effective amount is from 0.1 mg/m 2 to 2.0 mg/m 2.
  • the methods described herein further comprise imaging PSMA expression by the cancer.
  • the step of imaging occurs before the step of administering.
  • the imaging is performed by imaging and the imaging is selected from the group consisting of SPECT imaging, PET imaging, IHC, and FISH.
  • the imaging is performed by SPECT imaging.
  • the step of imaging comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • the step of imaging comprises administering a PSMA ligand-imaging conjugate of the formula III or a pharmaceutically acceptable salt thereof, wherein R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein M is a cation of a radionuclide.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium. In some aspects of these embodiments, M in the conjugate, or a pharmaceutically acceptable salt thereof, is an isotope of technetium.
  • the PSMA ligand-imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the methods described herein further comprise determining the PSMA status of the patient by imaging.
  • the imaging is SPECT imaging.
  • the PSMA status of the patient correlates with a clinical benefit to the patient.
  • the clinical benefit is selected from the group consisting of inhibition of tumor growth, stable disease, a partial response, and a complete response.
  • the clinical benefit is stable disease.
  • the step of determining comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • the step of determining comprises administering a PSMA ligand-imaging conjugate of the formula III
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is an isotope of technetium.
  • the PSMA ligand- imaging conjugate is of the formula Ila
  • the PSMA ligand-imaging conjugate is of the formula Ilia
  • the methods described herein further comprise determining the PSA level in a patient.
  • the PSA level can be measured from serum.
  • the methods described herein further comprise determining the PSA level in a patent prior to administering PSMA ligand-tubulysin compound I, and after administering PSMA ligand-tubulysin compound I.
  • the PSA level can be correlated with a clinical outcome.
  • the clinical outcome can be identified by a >30% decline in PSA.
  • the clinical outcome can be identified by a >50% decline in PSA.
  • a method for treating a cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • the cancer is selected from the group consisting of a glioma, a carcinoma, a sarcoma, a lymphoma, a melanoma, a mesothelioma, a nasopharyngeal carcinoma, a leukemia, an adenocarcinoma, and a myeloma.
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non- small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic leukemia, acute leukemia, lymphocytic lymphomas, pleural mes
  • the therapeutically effective amount is from about 0.1 mg/m 2 to about 100.0 mg/m 2 , or about 0.1 mg/m 2 to about
  • the therapeutically effective amount is from 0.1 mg/m 2 to 100.0 mg/m 2 , or 0.1 mg/m 2 to 90.0 mg/m 2 , or 0.1 mg/m 2 to 80.0 mg/m 2 , or 0.1 mg/m 2 to 70.0 mg/m 2 , or 0.1 mg/m 2 to 60.0 mg/m 2 , or 0.1 mg/m 2 to 50.0 mg/m 2 , or 0.1 mg/m 2 to 40.0 mg/m 2 , or 0.1 mg/m 2 to 35.0 mg/m 2 , or 0.1 mg/m 2 to 30.0 mg/m 2 , or 0.1 mg/m 2 to 25.0 mg/m 2 , or 0.1 mg/m 2 to 24.5 mg/m 2 , or 0.1 mg/m 2 to 24.0 mg/m 2 , or 0.1 mg/m 2 to 23.5 mg/m 2 , or 0.1 mg/m 2 to 23.0 mg/m 2 , or 0.1 mg/m 2 to
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • step of determining comprises administering to the patient a PSMA ligand-imaging conjugate of the formula II
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein M is a cation of a radionuclide.
  • M in the conjugate, or a pharmaceutically acceptable salt thereof is selected from the group consisting of an isotope of gallium, an isotope of indium, an isotope of copper, an isotope of technetium, and an isotope of rhenium.
  • a method of treating a cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • systemic treatment is selected from the group consisting of palifosfamide, 5-fluorouracil, capecitabine, pemetrexed, cisplatin, carboplatin, gemcitabine, paclitaxel, vinorelbine, eribulin, docetaxel, cyclophosphamide, doxorubicin, regorafinib, and combinations thereof.
  • the cancer is selected from the group consisting of lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non-small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic leukemia, acute leukemia, lymphocytic lymphomas, pleural
  • parenteral dosage form is selected from the group consisting of intradermal, subcutaneous, intramuscular, intraperitoneal, intravenous, and intrathecal.
  • the therapeutically effective amount is from 0.1 mg/m 2 to about 100.0 mg/m 2 , or about 0.1 mg/m 2 to about 90.0 mg/m 2 , or about 0.1 mg/m 2 to about 80.0 mg/m 2 , or about 0.1 mg/m 2 to about 70.0 mg/m 2 , or about 0.1 mg/m 2 to about 60.0 mg/m 2 , or about 0.1 mg/m 2 to about 50.0 mg/m 2 , or 0.1 mg/m 2 to about 40.0 mg/m 2 , or about 0.1 mg/m 2 to about 35.0 mg/m 2 , or about 0.1 mg/m 2 to about 30.0 mg/m 2 , or about 0.1 mg/m 2 to about 25.0 mg/m 2 , or 0.1 mg/m 2 to about 24.5 mg/m 2 , or about
  • 0.1 mg/m 2 to about 24.0 mg/m 2 or about 0.1 mg/m 2 to about 23.5 mg/m 2 , or about 0.1 mg/m 2 to about 23.0 mg/m 2 , or about 0.1 mg/m 2 to about 22.5 mg/m 2 , or about 0.1 mg/m 2 to about 22.0 mg/m 2 , or about 0.1 mg/m 2 to about 21.5 mg/m 2 , or about 0.1 mg/m 2 to about 21.0 mg/m 2 , or about 0.1 mg/m 2 to about 20.0 mg/m 2 , or about 0.1 mg/m 2 to about 19.0 mg/m 2 , or about 0.1 mg/m 2 to about 18.0 mg/m 2 , or about 0.1 mg/m 2 to about 17.0 mg/m 2 , or about 0.1 mg/m 2 to about 16.0 mg/m 2 , or about 0.1 mg/m 2 to about 15.0 mg/m 2 , or about 0.1 mg/m 2 to about 14.0 mg/m 2 , or about 0.1
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to the conjugate.
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherein a radionuclide is bound to a conjugate.
  • R' is hydrogen, or R' is selected from the group consisting of alkyl, aminoalkyl, carboxyalkyl, hydroxyalkyl, heteroalkyl, aryl, arylalkyl and heteroarylalkyl, each of which is optionally substituted, and wherin M is a cation of a radionuclide.
  • a method of treating a PSMA expressing cancer in a patient in need of such treatment comprising, administering to the patient a therapeutically effective amount of a PSMA ligand-tubulysin compound I
  • the present disclosure provides use of a PSMA ligand- tubulysin compound I
  • Fig. 1A shows a PET bone scan of a tumor lesion
  • Fig. IB shows a PSMA-imaging conjugate Ilia ( 99m Tc-EC0652) scan of the tumor lesion shown in Fig. 1A.
  • PSMA-imaging conjugate Ilia 99m Tc-EC0652 showed good uptake in the tumor lesion.
  • PSMA functionally active PSMA
  • PSMA ligands are well known to those skilled in the art such as those described in US patent publication no. US 2010/0324008 Al, incorporated herein by reference.
  • clinical benefit means a response of a patient to treatment with PSMA ligand-tubulysin compound I where the response includes overall survival of the patient, ability to receive four or more cycles of therapy (e.g., four weeks of therapy) with PSMA ligand-tubulysin compound I, inhibition of tumor growth, stable disease, a partial response, and/or a complete response, among other clinical benefits defined by the Food and Drug Administration in the United States of America.
  • inhibiting tumor growth means reduction in tumor size, complete disappearance of a tumor, or growth of a patient tumor of less than 30% over the course of therapy with PSMA ligand-tubulysin compound I.
  • stable disease means no material progression of disease in a patient over the course of therapy with PSMA ligand-tubulysin compound I.
  • a partial response means a decrease in tumor size of 30% or greater in a patient treated with PSMA ligand-tubulysin compound I.
  • a complete response means the disappearance of detectable disease in a patient treated with PSMA ligand-tubulysin compound I.
  • prior treatment means the patient has been treated with at least one prior treatment known in the art. It will be appreciated that a prior treatment can be any treatment known to those of skill in the art, including, but not limited, chemotherapeutic agent, surgery, radiation therapy, immunotherapy, photodynamic therapy, stem cell therapy, hyperthermia, and the like.
  • Prior treatments can include systemic treatments including, but not limited to treatment with palifosfamide, 5-fluorouracil, capecitabine, pemetrexed, cisplatin, carboplatin, gemcitabine, paclitaxel, vinorelbine, eribulin, docetaxel, cyclophosphamide, doxorubicin, regorafinib, and combinations thereof.
  • alkyl includes a chain of carbon atoms, which is optionally branched. It will be further understood that in certain embodiments, alkyl is advantageously of limited length, including Ci-C 24 , Ci-Ci 2 , Ci-C 8 , Ci-C 6 , and Ci-C 4 .
  • alkyl groups including Q-Cg, Ci-C 6 , and Ci-C 4 may be referred to as lower alkyl. It is appreciated herein that shorter alkyl, alkenyl, and/or alkynyl groups may add less lipophilicity to the compound and accordingly will have different pharmacokinetic behavior. In embodiments of the invention described herein, it is to be understood, in each case, that the recitation of alkyl refers to alkyl as defined herein, and optionally lower alkyl.
  • Illustrative alkyl groups include, but not limited to, methyl, ethyl, n- propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, 3-pentyl, neopentyl, hexyl, heptyl, octyl, and the like.
  • a "carboxyalkyl” group includes a
  • a "alkyl” group as described herein with a “carboxy” group includes a combination of an “alkyl” group as described herein with a “carboxy” group.
  • a “hydroxyalkyl” group includes a combination of an “alkyl” group as described herein with a “hydroxy” group.
  • a “aminoalkyl” group includes a combination of an “alkyl” group as described herein with a “amino” group.
  • heteroalkyl includes a chain of atoms that includes both carbon and at least one heteroatom, and is optionally branched.
  • Illustrative heteroatoms include nitrogen, oxygen, and sulfur. In certain variations, illustrative heteroatoms also include phosphorus, and selenium.
  • aryl includes monocyclic and polycyclic aromatic carbocyclic groups having from 6 to 14 ring carbon atoms, each of which may be optionally substituted.
  • Illustrative aromatic carbocyclic groups described herein include, but are not limited to, phenyl, naphthyl, and the like.
  • heteroaryl includes aromatic heterocyclic groups, having from 5 to 10 ring atoms, each of which may be optionally substituted.
  • Illustrative aromatic heterocyclic groups include, but are not limited to, pyridinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl, quinolinyl, quinazolinyl, quinoxalinyl, thienyl, pyrazolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, triazolyl, benzimidazolyl, benzoxazolyl, benzthiazolyl,
  • heteroarylalkyl includes a combination of an “alkyl” group as described herein with a
  • arylalkyl includes a combination of an "alkyl” group as described herein with a “aryl” group described herein, for example a benzyl group.
  • optionally substituted includes the replacement of hydrogen atoms with other functional groups on the radical that is optionally substituted.
  • Such other functional groups illustratively include, but are not limited to, amino, hydroxyl, halo, thiol, alkyl, haloalkyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, nitro, sulfonic acids and derivatives thereof, carboxylic acids and derivatives thereof, and the like.
  • any of amino, hydroxyl, thiol, alkyl, haloalkyl, heteroalkyl, aryl, arylalkyl, arylheteroalkyl, heteroaryl, heteroarylalkyl, heteroarylheteroalkyl, and/or sulfonic acid is optionally substituted.
  • the term "administering" as used herein includes all means of introducing the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein to the patient, including, but not limited to, oral (po), intravenous (iv), intramuscular (im), subcutaneous (sc), transdermal, inhalation, buccal, ocular, sublingual, vaginal, rectal, and the like.
  • the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein may be administered in unit dosage forms and/or formulations containing conventional nontoxic pharmaceutically-acceptable carriers, adjuvants, and vehicles.
  • a "patient” can be administered the PSMA ligand- tubulysin compound or PSMA ligand-imaging conjugates described herein, and can be human or, in the case of veterinary applications, can be a laboratory, agricultural, domestic, or wild animal.
  • the patient can be a human, a laboratory animal such as a rodent (e.g.
  • the cancers described herein can be a cancer cell population that is tumorigenic, including benign tumors and malignant tumors, or the cancer can be non- tumorigenic.
  • the cancer can arise spontaneously or by such processes as mutations present in the germline of the patient or somatic mutations, or the cancer can be chemically-, virally-, or radiation-induced.
  • Cancers applicable to the invention described herein include, but are not limited to, a glioma, a carcinoma, a sarcoma, a lymphoma, a melanoma, a mesothelioma, a nasopharyngeal carcinoma, a leukemia, an adenocarcinoma, and a myeloma.
  • the cancers can be lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head, cancer of the neck, cutaneous melanoma, intraocular melanoma uterine cancer, ovarian cancer, endometrial cancer, rectal cancer, stomach cancer, colon cancer, breast cancer, triple negative breast cancer, metastatic breast cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, non-small cell lung cancer, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, chronic leukemia, acute leukemia, lymphocytic lymphomas, pleural mesothelioma, cancer of the bladder, Burkitt's lymph
  • PSMA ligand-imaging conjugates detectable by PET imaging, SPECT imaging, and the like can be used.
  • the exact manner of imaging is not limited to the imaging agents described herein.
  • PSMA ligand-imaging conjugates useful for imaging described herein, including those described by formulas and the agents useful for PET imaging, SPECT imaging, etc. are referred to as "PSMA ligand-imaging conjugates.”
  • the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein bind to expressed PSMA on cancer cells.
  • the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates are capable of differentially binding to PSMA on cancer cells compared to normal cells due to preferential expression (or over-expression) of PSMA on the cancer cells.
  • pharmaceutically acceptable salts of the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein are provided.
  • Pharmaceutically acceptable salts of the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein include acid addition and base salts thereof.
  • Suitable acid addition salts are formed from acids which form non-toxic salts.
  • Illustrative examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tos
  • Suitable base salts of the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein are formed from bases which form non-toxic salts.
  • bases which form non-toxic salts.
  • Illustrative examples include the arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
  • Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • the PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein may be administered as a formulation in association with one or more pharmaceutically acceptable carriers.
  • the carriers can be excipients.
  • the choice of carrier will to a large extent depend on factors such as the particular mode of administration, the effect of the carrier on solubility and stability, and the nature of the dosage form.
  • Pharmaceutical compositions suitable for the delivery of PSMA ligand-tubulysin compound and PSMA ligand-imaging conjugates described herein and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington: The Science & Practice of Pharmacy, 21th Edition (Lippincott Williams & Wilkins, 2005), incorporated herein by reference.
  • a pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, and combinations thereof, that are physiologically compatible.
  • the carrier is suitable for parenteral administration.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. Supplementary active compounds can also be incorporated into compositions of the invention.
  • liquid formulations may include suspensions and solutions.
  • Such formulations may comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose or a suitable oil, and one or more emulsifying agents and/or suspending agents.
  • Liquid formulations may also be prepared by the reconstitution of a solid.
  • an aqueous suspension may contain the active materials in admixture with appropriate excipients.
  • excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally-occurring phosphatide, for example, lecithin; a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate; a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadecaethyleneoxycetanol; a condensation product of ethylene oxide with a partial ester derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate; or a condensation product of ethylene oxide with a partial ester derived from fatty acids and hexitol anhydrides, for example,
  • dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Additional excipients, for example, coloring agents, may also be present.
  • Suitable emulsifying agents may be naturally-occurring gums, for example, gum acacia or gum tragacanth; naturally-occurring phosphatides, for example, soybean lecithin; and esters including partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan mono-oleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride can be included in the composition.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin.
  • Illustrative formats for oral administration include tablets, capsules, elixirs, syrups, and the like.
  • routes of administration and/or whether the PSMA ligand-tubulysin compound and/or PSMA ligand-imaging conjugates are administered locally or systemically a wide range of permissible dosages are contemplated herein, including doses falling in the range from about 1 ⁇ g/kg to about 1 g/kg.
  • the dosages may be single or divided, and may administered according to a wide variety of protocols, including q.d., b.i.d., t.i.d., or even every other day, biweekly (b.i.w.), once a week (QW), once a month, once a quarter, and the like.
  • q.d. b.i.d.
  • t.i.d. t.i.d.
  • QW once a week
  • the therapeutically effective amounts described herein correspond to the instance of administration, or alternatively to the total daily, weekly, monthly, or quarterly dose, as determined by the dosing protocol.
  • a PSMA ligand-tubulysin compound or a PSMA ligand-imaging conjugate as described herein may be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable routes for such parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, epidural, intracerebroventricular, intraurethral, intrasternal, intracranial, intratumoral, intramuscular and subcutaneous delivery.
  • Suitable means for parenteral administration include needle (including microneedle) injectors, needle- free injectors and infusion techniques.
  • parenteral formulations are typically aqueous solutions which may contain carriers or excipients such as salts, carbohydrates and buffering agents (preferably at a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • a suitable vehicle such as sterile, pyrogen-free water.
  • any of the liquid formulations described herein may be adapted for parenteral administration of the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates described herein.
  • parenteral formulations under sterile conditions may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
  • solubility of a PSMA ligand-tubulysin compound or a PSMA ligand-imaging conjugate used in the preparation of a parenteral formulation may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents.
  • formulations for parenteral administration may be formulated for immediate and/or modified release.
  • active agents of the invention i.e., the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates
  • the active PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates can be prepared with carriers that will protect the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugate against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid and polylactic, polyglycolic copolymers (PGLA). Methods for the preparation of such formulations are generally known to those skilled in the art.
  • the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates described herein or compositions comprising the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates may be continuously administered, where appropriate.
  • kits are provided. If a combination of active PSMA ligand- tubulysin compound and PSMA ligand-imaging conjugates is to be administered, two or more pharmaceutical compositions may be combined in the form of a kit suitable for sequential administration or co-administration of the compositions.
  • a kit comprises two or more separate pharmaceutical compositions, at least one of which contains a PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugate described herein, and means for separately retaining the compositions, such as a container, divided bottle, or divided foil packet.
  • compositions comprising one or more of the PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugates described herein, in containers having labels that provide instructions for use of the PSMA ligand-tubulysin compound or PSMA ligand- imaging conjugates for patient selection and/or treatment are provided.
  • sterile injectable solutions can be prepared by incorporating the active agent in the required amount in an appropriate solvent with one or a combination of ingredients described above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active PSMA ligand-tubulysin compound or PSMA ligand-imaging conjugate into a sterile vehicle which contains a dispersion medium and any additional ingredients of those described above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof, or the ingredients may be sterile-filtered together.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • PSMA ligand-tubulysin compound I can be administered as single doses, or the doses can be divided and administered as a multiple-dose daily regimen.
  • a staggered regimen for example, one to five days per week can be used as an alternative to daily treatment, and for the purpose of the methods described herein, such intermittent or staggered daily regimen is considered to be equivalent to every day treatment and is contemplated.
  • the patient is treated with multiple injections of PSMA ligand-tubulysin compound I to treat the cancer.
  • the patient is injected multiple times (preferably about 2 up to about 50 times) with PSMA ligand-tubulysin compound I, for example, at 12-72 hour intervals or at 48-72 hour intervals. Additional injections of PSMA ligand-tubulysin compound I can be administered to the patient at an interval of days or months after the initial injections(s) and the additional injections can prevent recurrence of the cancer.
  • PSMA ligand-tubulysin compound I is administered in a single daily dose administered five days a week, in weeks 1, 2, and 3 of each 4 week cycle, with no dose administered in week 4.
  • PSMA ligand-tubulysin compound I is administered in a single daily dose administered three days a week, of weeks 1, and 3 of each 4 week cycle, with no dose administered in weeks 2 and 4.
  • PSMA ligand-tubulysin compound I is administered biweekly on weeks 1 and 2, i.e. on days 1, 4, 8, 11, of a 3-week cycle.
  • PSMA ligand-tubulysin compound I is administered once weekly on weeks 1 and 2, i.e. days 1 and 8 of a 3-week cycle.
  • PSMA ligand-tubulysin compound I is administered as an IV bolus injection at least 4 days after an imaging conjugate is administered, and on a schedule of once weekly (QW) on Day 1 and Day 8 of a 21 -day cycle.
  • PSMA ligand-tubulysin compound I is administered as an IV bolus injection on a schedule of once weekly (QW) on Day 1 and Day 8 of a 21 -day cycle.
  • the unitary daily dosage of the PSMA ligand-tubulysin compound I can vary significantly depending on the patient condition, the cancer being treated, the route of administration of the PSMA ligand-tubulysin compound I and tissue distribution, and the possibility of co-usage of other therapeutic treatments, such as radiation therapy or additional drugs in combination therapies.
  • the effective amount to be administered to a patient is based on body surface area, mass, and physician assessment of patient condition.
  • Therapeutically effective doses can range, for example, from about 0.1 mg/m 2 to about 100.0 mg/m 2.
  • the therapeutically effective doses described herein also include ranges of about 0.1 mg/m 2 to about 90.0 mg/m 2 , about 0.1 mg/m 2 to about 80.0 mg/m 2 , about 0.1 mg/m 2 to about 70.0 mg/m 2 , about 0.1 mg/m 2 to about 60.0 mg/m 2 , about 0.1 mg/m 2 to about 50.0 mg/m 2 , 0.1 mg/m 2 to about 40.0 mg/m 2 , about 0.1 mg/m 2 to about 35.0 mg/m 2 , about
  • 0.1 mg/m 2 to about 30.0 mg/m 2 about 0.1 mg/m 2 to about 25.0 mg/m 2 , 0.1 mg/m 2 to about 24.5 mg/m 2 , about 0.1 mg/m 2 to about 24.0 mg/m 2 , about 0.1 mg/m 2 to about 23.5 mg/m 2 , about 0.1 mg/m 2 to about 23.0 mg/m 2 , about 0.1 mg/m 2 to about 22.5 mg/m 2 , about 0.1 mg/m 2 to about 22.0 mg/m 2 , about 0.1 mg/m 2 to about 21.5 mg/m 2 , about 0.1 mg/m 2 to about 21.0 mg/m 2 , about
  • 0.1 mg/m 2 to about 20.0 mg/m 2 about 0.1 mg/m 2 to about 19.5 mg/m 2 , about 0.1 mg/m 2 to about 19.0 mg/m 2 , about 0.1 mg/m 2 to about 18.5 mg/m 2 , about 0.1 mg/m 2 to about 18.0 mg/m 2 , about 0.1 mg/m 2 to about 17.5 mg/m 2 , about 0.1 mg/m 2 to about 17.0 mg/m 2 , about 0.1 mg/m 2 to about 16.5 mg/m 2 , about 0.1 mg/m 2 to about 16.0 mg/m 2 , about 0.1 mg/m 2 to about 15.5 mg/m 2 , about 0.1 mg/m 2 to about 15.0 mg/m 2 , about 0.1 mg/m 2 to about 14.5 mg/m 2 , about 0.1 mg/m 2 to about 14.0 mg/m 2 , about 0.1 mg/m 2 to about 13.5 mg/m 2 , about 0.1 mg/m 2 to about
  • 0.1 mg/m 2 to about 11.5 mg/m 2 about 0.1 mg/m 2 to about 11.0 mg/m 2 , about 0.1 mg/m 2 to about 10.5 mg/m 2 , about 0.1 mg/m 2 to about 10.0 mg/m 2 , about 0.1 mg/m 2 to about 9.5 mg/m 2 , about 0.1 mg/m 2 to about 9.0 mg/m 2 , about 0.1 mg/m 2 to about 8.5 mg/m 2 , about 0.1 mg/m 2 to about 8.0 mg/m 2 , about 0.1 mg/m 2 to about 7.5 mg/m 2 , about 0.1 mg/m 2 to about 7.0 mg/m 2 , about 0.1 mg/m 2 to about 6.5 mg/m 2 , about 0.1 mg/m 2 to about 6.0 mg/m 2 , about 0.1 mg/m 2 to about 5.5 mg/m 2 , about 0.1 mg/m 2 to about 5.0 mg/m 2 , about 0.1 mg/m 2 to about 4.5 mg/m 2 , about
  • 1.0 mg/m 2 to about 11.5 mg/m 2 about 1.0 mg/m 2 to about 11.0 mg/m 2 , about 1.0 mg/m 2 to about 10.5 mg/m 2 , about 1.0 mg/m 2 to about 10.0 mg/m 2 , about 1.0 mg/m 2 to about 9.5 mg/m 2 , about 1.0 mg/m 2 to about 9.0 mg/m 2 , about 1.0 mg/m 2 to about 8.5 mg/m 2 , about 1.0 mg/m 2 to about 8.0 mg/m 2 , about 1.0 mg/m 2 to about 7.5 mg/m 2 , about 1.0 mg/m 2 to about 7.0 mg/m 2 , about 1.0 mg/m 2 to about 6.5 mg/m 2 , about 1.0 mg/m 2 to about 6.0 mg/m 2 , about 1.0 mg/m 2 to about 5.5 mg/m 2 , about 1.0 mg/m 2 to about 5.0 mg/m 2 , about 1.0 mg/m 2 to about 4.5 mg/m 2 , about
  • the therapeutically effective dose may vary within the various ranges provided above based on the factors noted above.
  • the therapeutically effective dose for any particular patient or group of patients may be any number value between about 0.1 mg/m 2 and about 100.0 mg/m 2 , including but not limited to 1.0 mg/m 2 , 1.5, mg/m 2 , 2.0 mg/m 2 , 2.5 mg/m 2 , 3.0 mg/m 2 , 3.5 mg/m 2 , 4.0 mg/m 2 , 4.5 mg/m 2 , 5.0 mg/m 2 , 5.5 mg/m 2 , 6.0 mg/m 2 , 6.5 mg/m 2 , 7.0 mg/m 2 , 7.5 mg/m 2 , 8.0 mg/m 2 , 8.5 mg/m 2 , 9.0 mg/m 2 , 9.5 mg/m 2 , 10.0 mg/m 2 , 10.5 mg/m 2 , 11.0 mg/m 2 , 11.5 mg/m 2 ,
  • the total dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein.
  • the PSMA ligand-imaging conjugates and PSMA ligand-tubulysin compound described herein may contain one or more chiral centers, or may otherwise be capable of existing as multiple stereoisomers. Accordingly, it is to be understood that the present invention includes pure stereoisomers as well as mixtures of stereoisomers, such as enantiomers, diastereomers, and enantiomerically or diastereomerically enriched mixtures.
  • the PSMA ligand-imaging conjugates and PSMA ligand-tubulysin compound described herein may be capable of existing as geometric isomers. Accordingly, it is to be understood that the present invention includes pure geometric isomers or mixtures of geometric isomers.
  • PSMA ligand-imaging conjugates and PSMA ligand-tubulysin compound described herein may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention.
  • the PSMA ligand-imaging conjugates and PSMA ligand-tubulysin compound described herein may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • compositions and/or dosage forms for administration of PSMA ligand-tubulysin compound I are prepared from PSMA ligand-tubulysin compound I with a purity of at least about 90%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99%, or about 99.5%.
  • compositions and or dosage forms for administration of PSMA ligand-tubulysin compound I are prepared from PSMA ligand- tubulysin compound I with a purity of at least 90%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%, or at least 99.5%.
  • compositions and/or dosage forms for administration of the PSMA ligand-imaging conjugate are prepared from the PSMA ligand-imaging conjugate with a purity of at least about 90%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99%, or about 99.5%.
  • compositions and or dosage forms for administration of the PSMA ligand-imaging conjugate are prepared from the PSMA ligand- imaging conjugate with a purity of at least 90%, or at least 95%, or at least 97%, or at least 98%, or at least 99%, or at least 99.5%.
  • compositions and/or dosage forms for administration of radiolabeled PSMA ligand-imaging conjugate are prepared from the PSMA ligand-imaging conjugate with a radiochemical purity of at least about 90%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99%, or about 99.5%.
  • compositions and or dosage forms for administration of the PSMA ligand-imaging conjugate are prepared from the PSMA ligand-imaging conjugate with a purity of at least 90%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%, or at least 99.5%.
  • PSMA ligand-tubulysin compound I or the PSMA ligand-imaging conjugates described herein may be measured using any conventional technique, including various chromatography or spectroscopic techniques, such as high pressure or high performance liquid chromatography (HPLC), nuclear magnetic resonance spectroscopy, TLC, UV absorbance spectroscopy, fluorescence spectroscopy, and the like.
  • HPLC high pressure or high performance liquid chromatography
  • TLC nuclear magnetic resonance spectroscopy
  • UV absorbance spectroscopy fluorescence spectroscopy
  • the PSMA ligand-tubulysin compound or PSMA ligand- imaging conjugate described herein is provided in a sterile container or package.
  • a clinical benefit of the patient to treatment with PSMA ligand-tubulysin compound I can be characterized utilizing Response Evaluation Criteria in Solid Tumors (RECIST) criteria.
  • the criteria have been adapted from the original WHO Handbook (3), taking into account the measurement of the longest diameter for all target lesions: complete response, (CR)— the disappearance of all target lesions; partial response (PR)— at least a 30% decrease in the sum of the longest diameter of target lesions, taking as reference the baseline sum longest diameter; stable disease (SD)— neither sufficient shrinkage to qualify for partial response nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum longest diameter since the treatment started; progressive disease (PD)— at least a 20% increase in the sum of the longest diameter of target lesions, taking as reference the smallest sum longest diameter recorded since the treatment started or the appearance of one or more new lesions.
  • overall disease response rate is a clinical benefit and is calculated as the percent of patients who achieve a best response of CR or PR.
  • Overall disease control rate (DCR) can be another clinical benefit and is calculated as the percent of patients who achieve a best response of CR, PR, or SD.
  • overall survival is the time to death for a given patient defined as the number of days from the first day the patient received protocol treatment (C1D1) to the date of the patient's death. All events of death can be included, regardless of whether the event occurred while the patient was still taking the study drug or after the patient discontinued the study drug. If a patient has not died, then the data can be censored at the last study visit, or the last contact date, or the date the patient was last known to be alive, whichever is last.
  • C1D1 patient received protocol treatment
  • a clinical benefit of the patient as a result of treatment with PSMA ligand- tubulysin compound I can be characterized as inhibition of tumor growth which can be identified in a patient through, for example, follow-up imaging of the patient's cancer after treatment with PSMA ligand-tubulysin compound I.
  • inhibition of tumor growth can be characterized by measuring the size of tumors in a patient after administration of PSMA ligand-tubulysin compound I according to any of the imaging techniques described herein, where the inhibition of tumor growth is indicated by a stable tumor size, or by a reduction in tumor size. It will be appreciated that the identification of inhibition of tumor growth can be accomplished using a variety of techniques, and is not limited to the imaging methods described herein (e.g CT, MRI, PET imaging, SPECT imaging or chest x-ray).
  • a method is provided of determining whether PSMA ligand- tubulysin compound I is indicated for the treatment of a patient with cancer, the method comprising the step of determining the PSMA status in a patient with cancer wherein PSMA ligand-tubulysin compound I is indicated for the treatment of the patient if the PSMA status of the patient is positive.
  • a method is provided of assessing whether PSMA ligand-tubulysin compound I is indicated for the treatment of a patient with one of the cancers described herein.
  • the method comprises the steps of visually determining PSMA status in the patient wherein PSMA status is based on a imaging tumors that are PSMA positive in the patient, and wherein the PSMA ligand-tubulysin compound I is indicated for the treatment of the patient when the PSMA status of the patient is positive.
  • a clinical benefit of PSMA ligand-tubulysin compound I treatment is indicated.
  • the clinical benefit to the patient can be overall survival of the patient, ability to receive four or more cycles of therapy with PSMA ligand-tubulysin compound I, inhibition of tumor growth, stable disease, a partial response of the patient to therapy, a complete response of the patient to therapy, disease control (i.e., the best result obtained is a complete response, a partial response, or stable disease), and/or overall disease response (i.e., the best result obtained is a complete response or a partial response).
  • the clinical benefit for a patient being treated for pleural mesothelioma or adenocarcinoma is stable disease.
  • a clinical outcome associated with administration of PSMA ligand-tubulysin compound I can be identified by a number of measures or markers known in the art.
  • a patient's PSA level can be measured to determine a clinical outcome.
  • the methods described herein further comprise determining the PSA level in a patient.
  • the PSA level in a patient can be determined by any means known to one of skill in the art, including, by measuring PSA in serum taken from a patient.
  • Assays useful in connection with measuring or determining PSA in a patient sample such as seminal fluid, seminal fluid coagulum, whole blood, serum, urine, and the like, are well known in the art.
  • the methods described herein further comprise determining the PSA level in a patent prior to administering PSMA ligand-tubulysin compound I, and after administering PSMA ligand-tubulysin compound I.
  • the PSA level can be correlated with a clinical outcome.
  • the clinical outcome can be identified by a >20% decline in PSA. In some embodiments, the clinical outcome can be identified by a >30% decline in PSA.
  • the clinical outcome can be identified by a >40% decline in PSA. In some embodiments, the clinical outcome can be identified by a >50% decline in PSA. In some embodiments, the clinical outcome can be identified by a >60% decline in PSA.
  • the methods described herein include the following examples.
  • the examples further illustrate additional features of the various embodiments of the invention described herein.
  • the examples are illustrative and are not to be construed as limiting other embodiments of the invention described herein.
  • other variations of the examples are included in the various embodiments of the invention described herein.
  • PSMA ligand-tubulysin compound I was prepared according to the methods described in United States Patent Publication No. WO2014/078484, incorporated herein by reference. Specifically, PSMA ligand-tubulysin compound I is prepared according to the following procedure. a. Synthesis of 3-nitro-2-disulfenylethanol 2.
  • TES protected dipeptide was dissolved in 100 mL THF (anhydrous, inhibitor- free), cooled to -45 °C, and stirred at -45 °C for 15 minutes before adding KHMDS (0.5 M in toluene, 61 mL, 1.05 equiv.), drop-wise. After the addition of KHMDS was finished, the reaction was stirred at -45 °C for 20 minutes, and chloromethyl butyrate (4.4 mL, 1.1 equiv.) was added. The reaction mixture was stirred at -45 °C for another 20 minutes. The reaction was quenched with 25 mL MeOH and warmed to room temperature.
  • alkylated dipeptide 9 (26.97 mmol.), N-methyl pipecolinate (MEP) (5.51 g, 1.4 equiv.) and pentafluorophenol (7.63 g, 1.5 equiv.) were added to a 300 mL hydrogenation flask. NMP (115 mL) was then added, followed by EDC (7.78 g, 1.5 equiv.). The mixture was stirred at room temperature for overnight. 16.5 g of alkylated dipeptide 9 was dissolved in 16.5 mL NMP, transferred the solution into the hydrogenation flask, washed the residual 9 with 8 mL NMP, and transferred into the hydrogenation flask.
  • Methyl ester 10 (6.9 g, 9.7 mmol) was dissolved in 1,2-dichloroethane (193 mL) and added to a round bottomed flask, equipped with a stir bar and condenser. To this solution was added trimethyltin hydroxide (24.6 g, 14 eq.). The mixture was heated at 70 °C for 5 hours. LC-MS analysis indicated that the desired product had been formed and ⁇ 15 % of starting methyl ester 10 remained. The reaction was cooled in an ice bath for 30 minutes. The resulting precipitate was then removed by filtration. The filtrate was stored overnight at -20 °C. The filtrate was then divided into two portions and each was subjected the chromatography procedure which follows.
  • tripeptide acid 11 (3.9 g, 5.6 mmol) was dissolved in anhydrous THF (23 mL). To this solution was added 3 HF'TEA complex (1.8 mL, 2 eq.). The reaction was stirred at room temperature for 1 hour. LC-MS analysis indicated complete conversion to the desired des-TES product 12. The solvent was removed under reduced pressure and the residue was placed on the high vacuum for 40 minutes. The resulting residue was then dissolved in pyridine (26 mL), and acetic anhydride (7.9 mL, 15 eq.) and DMAP (25 mg) were added. The reaction was stirred at room temperature for 1 hour.
  • the activated Boc-Tut- fragment 6 (2.63 g, 4.42 mmol, 1.1 equiv) was treated with TFA/CH 2 C1 2 (42 mL; 1: 1) and stirred for 30 minutes.
  • LC-MS analysis (X-Bridge shield RP18, 3.5 ⁇ column; gradient 10% to 100% acetonitrile in 6 min, pH 7.4 buffer) confirmed the product formation.
  • TFA was removed under reduced pressure, co-evaporated with CH 2 CI 2 (3 x 30 mL) and activated Tut-derivative 14 was dried under high vacuum for 18h.
  • H-Glu(OtBu)-OtBu HCl (101) (4.83g, 16.3 mmol, available from Sigma- Aldrich) and 4-nitrophenyl chloroformate (102) (3.47g, 17.2 mmol, available from Sigma- Aldrich) were dissolved in dichloromethane (50mL) and stirred in an ice bath under argon. Diisopropylethylamine (6.28mL, 36.1 mmol) was added slowly, dropwise and the reaction mixture was stirred in the ice bath for 5 min, then warmed to room temperature and stirred for 30 min.
  • Boc-4-aminomethylphenylacetic acid (106) (2.00g, 7.5 mmol, available from VWR) dissolved in a solution of trifluoroacetic acid (9.75mL) and triisopropylsilane (0.25mL) and stirred at room temperature for 30 min, then concentrated under reduced pressure and coevaporated with dichloromethane (3x), then placed under vacuum, to yield 4- aminomethylphenylacetic acid (107) (quantitative yield).
  • k Preparation of compound 108.
  • H-Cys(4-methoxytrityl)-2-chlorotrityl-resin (0.87 mmol) was loaded and washed with isopropyl alcohol (3xl0mL) followed by dimethylformamide (3xl0mL). To the vessel was then introduced Fmoc-Asp(OtBu)-OH (2.0 equiv) in
  • a peptide synthesis vessel 109 (0.18 mmol) was loaded and washed with isopropyl alcohol (3xl0mL) followed by dimethylformamide (3xl0mL). Fmoc deprotection was carried out using 20% piperidine in dimethylformamide (3x10 mL). Kaiser tests were performed to assess reaction completion. To the vessel was then introduced 108 (1.2 equiv) in
  • Peptide was cleaved from the resin using a cleavage mixture consisting of dithiothreitol (114mg, 0.74 mmol) dissolved in a solution of trifluoroacetic acid (19mL), H 2 0 (0.5mL), triisopropylsilane (0.5mL).
  • a cleavage mixture consisting of dithiothreitol (114mg, 0.74 mmol) dissolved in a solution of trifluoroacetic acid (19mL), H 2 0 (0.5mL), triisopropylsilane (0.5mL).
  • One-third of the cleavage mixture was introduced and argon was bubbled for 30 min.
  • the cleavage mixture was drained into a clean flask.
  • the resin was bubbled 2 more times with more cleavage mixture, for 30 min each, and drained into a clean flask.
  • PSMA imaging conjugate Ila was prepared according to the following scheme as taught in US patent publication number US20100324008 Al, which is incorporated herein by reference. Specifically, PSMA imaging conjugate Ila was prepared according to the following method.
  • PSMA imaging conjugate Ila was synthesized using standard
  • Fmoc fluorenylmethyloxycarbonyl
  • SPPS solid phase peptide synthesis
  • 1.0 M NaOH and 0.2 M HCI were prepared and sparged with nitrogen for pH adjustment of the formulation and for preparation of the stannous chloride stock solution.
  • 2000 mL of deoxygenated WFI was added to a 5L jacketed formulation vessel which was connected to a chiller.
  • the chiller solution was set at 5 °C and circulation was maintained throughout the compounding and filtration process.
  • 88.6 g of sodium gluconate and 1063 mg of EDTA disodium dihydrate were weighed and transferred to the formulation vessel and dissolved.
  • a stannous chloride stock solution at a concentration of lOmg/mL was made using the previously prepared 0.2 M HCI.
  • a PSMA imaging conjugate Ila kit vial was removed from the refrigerator and allowed to warm to room temperature (17-27 °C) for 15-30 min.
  • the vial was put into a suitable radioactive shielding container.
  • One to Two milliliter ( ⁇ 50 mCi) of 99m Tc pertechnetate injection was added to the vial using a lead shielded syringe.
  • equal volume of headspace was withdrawn in order to normalize the pressure inside the vial.
  • the vial was gently swirled to completely dissolve the powder and then allowed to stand at ambient temperature (17-27 °C) for 15 minutes.
  • 5-6 mL of 0.9% sodium chloride injection, USP was then added to the vial.
  • the labeled solution was stored at room temperature (17-27 °C) and used within 6 hours of preparation.
  • mCRPC metastatic, castration-resistant prostate cancer
  • EC0652 patients were also dosed with 99mTc-EC0652 and underwent a SPECT/CT scan (or SPECT scan if no SPECT/CT is available) to evaluate EC0652 as an imaging agent to identify PSMA expression. And this is an open-label, multicenter, Phase 1 study of ECl 169, conducted in 2 parts. Part A is a dose-escalation phase to determine the RP2 dose.
  • Prior dose escalation methodology for both schedules was based the continuous reassessment method (CRM), in which 1 patient is assigned to 1 dose level. Subsequent patients were to be enrolled upon the first observation of DLT, at which time enrollment to that dose level would be expanded to a maximum of 6 patients.
  • An alternative dose escalation methodology for both schedules was based upon the standard "3+3" approach, in which a minimum of 3 patients is enrolled to a given dose level. Following the first observation of a DLT, the dose level is then expanded to a maximum of 6 patients. A 28-day screening period will precede Cycle 1 Day 1. All patients receive a baseline
  • Radiographic assessment (computed tomography [CT] and bone scans) will be done at baseline and at the end of every 2nd cycle. Baseline PSA, serum testosterone, and CTC will be collected and repeated on Day 1 of every cycle. Patients may continue receiving EC 1169 until confirmed radiological disease
  • ICF informed consent form
  • Patients must be males > 18 years of age. 3. Patients must have histological, pathological, and/or cytological confirmation of prostate cancer.
  • mCRPC metastatic, castration-resistant prostate cancer
  • Documented progressive metastatic CRPC will be based on at least one of the following criteria:
  • PSA progression defined as 25% increase over a baseline value, such as > 2 ng/ml, with an increase in the absolute value of at least 2 ng/mL that is confirmed by another PSA level with a minimum of a 1 week interval.
  • Baseline is defined as the PSA nadir level since commencing most recent prior therapy.
  • Soft-tissue progression defined as an increase > 20% in the sum of the longest diameter (LD) of all target lesions based on the smallest sum LD since treatment started or the appearance of one or more new lesions, or an increase > 20% in the sum of the diameter (SOD; short axis for nodal lesions and long axis for non-nodal lesions) of all target lesions based on the smallest SOD, or the appearance of one or more new lesions, since the onset of the most recent prior therapy
  • Patients must have prior and/or ongoing androgen-deprivation therapy and/or a castrate level of serum testosterone ( ⁇ 50 ng/dL).
  • contraindication e.g. poor performance status, age, personal choice or drug intolerance.
  • Patients must have at least one lesion, such as a metastatic lesion, that can be followed for disease response assessment on baseline imaging obtained no more than 28 days prior to beginning study therapy.
  • Baseline and follow up radiological disease assessments must include bone scans performed with either Technetium-99m labeled diphosphonates or Fluorine- 18 sodium fluoride PET or PET/CT, as per the local standard of care for patients with prostate cancer.
  • CNS metastases that were symptomatic must have received therapy (surgery, radiotherapy (XRT), gamma knife) and been neurologically stable, asymptomatic, and off of steroids, such as corticosteroids for the purposes of maintaining neurologic activity.
  • Patients with epidural disease, canal disease and prior cord involvement are eligible if those areas have been treated, are stable, and not neurologically impaired.
  • the patients were off steroids at least 14 days before pre-registration.
  • Asymptomatic CNS metastatic disease without associated edema, shift, requirement for steroids or anti-seizure medications are eligible after discussion with the sponsor medical monitor.
  • baseline and subsequent radiological imaging included evaluation of the brain (MRI preferred or CT with contrast).
  • NCI National Cancer Institute
  • CCAE Common Terminology Criteria for Adverse Events
  • Bone marrow reserve Absolute neutrophil count (ANC) > 1.5 x 109/L. platelets > 100 x 10 9 /L. hemoglobin > 9 g/dL.
  • QTc Fridericia QTcF ⁇ 450 msec on at least 2 of 3 screening ECGs. On site determination of QTcF may be used for screening purposes.
  • LVEF Left ventricular ejection fraction
  • ALT aminotransferase
  • AST aspartate aminotransferase
  • Renal Serum/plasma creatinine ⁇ 1.5 x ULN, or for patients with serum/plasma creatinine > 1.5 ULN, creatinine clearance > 50 niL/min.
  • Any systemic anti-cancer therapy e.g., chemotherapy, immunotherapy or biological therapy [including monoclonal antibodies or experimental anti-cancer therapy] within 28 days prior to beginning study therapy.
  • Ongoing castrating therapy with a GnRH agonist or antagonist is mandatory to assure a castrate level of serum testosterone ⁇ 50 ng/dL, except in patients who have undergone an orchiectomy.
  • Bisphosphonates or denosumab continuation is permissible (i.e., no change for 30 days prior to Cycle 1 Day 1).
  • Patients who receive a dose of Compound I under another Endocyte protocol do not need a washout period for Compound I.
  • Malignancies that are expected to alter life expectancy e.g., NSCLC, etc.
  • that may interfere with disease assessment of prostate cancer e.g., lymphoma involving the periaortic nodes.
  • QTc heart rate
  • IV intravenous
  • 740-925 MBq 20-25 mCi
  • 99m Tc 99m Tc
  • uptake interval defined in the Investigators Imaging Operations Manual e.g., 3 hours post-injection based on estimates from nonclinical models
  • SPECT single-photon emission computed tomography
  • PSMA Imaging Agent Ila labeled with 20-25 mCi of technetium- 99 TM (PSMA Imaging Agent Ilia).
  • PSMA Imaging Agent Ilia PSMA Imaging Agent Ilia
  • Patients were subjected to SPECT/CT imaging of the region(s) known to contain target lesion(s) approximately 3-4 hours after injection of PSMA Imaging Agent Ilia ( 99m Tc-EC0652). For sites where SPECT/CT imaging is not available, SPECT imaging alone was carried out.
  • PSMA ligand-tubulysin compound I was administered (at least 4 days after the 99m Tc-EC0652 administration) as an IV bolus injection. Dosing regimen was once weekly (QW) on Day 1 and Day 8 of a 21 -day cycle. There was no maximum duration of treatment. Patients continued to receive PSMA ligand-tubulysin compound I until they are no longer clinically benefiting from therapy or have withdrawn from the study, contingent upon the availability of study drug. In some cases, PSMA ligand-tubulysin compound I was administered at least 4 days after
  • PSMA Imaging Agent Ilia was administered, as an intravenous bolus injection, ⁇ on Weeks 1 and 2 (i.e., on Days 1, 3, 5, 8, 10, 12 of a 3-week cycle) or once weekly on Weeks 1 and 2 of a 3-week schedule. II. Routes of Administration
  • PSMA Imaging Agent Ilia 99m Tc-EC0652
  • PSMA ligand-tubulysin compound I were administered via IV bolus injection.
  • IV Schedule #1 TIW Dosing Dose Escalation:
  • the starting dose of EC 1169 on Schedule 1 was 0.2 mg/m
  • the table below outlines the dose levels for PSMA ligand-tubulysin compound I for Schedule #1 TIW dosing, with up to 14 doses levels of PSMA ligand-tubulysin compound I planned. Should the MTD not be determined after escalation of PSMA ligand-tubulysin compound I to dose level 6, PSMA ligand-tubulysin compound I may continue to be dose escalated in 25% increments.
  • the starting dose of EC 1169 was 0.30 mg/m .
  • the table below outlines the dose levels for PSMA ligand-tubulysin compound I for Schedule #2 ⁇ dosing, with up to 14 doses levels of PSMA ligand-tubulysin compound I planned. Should the MTD not be determined after escalation of PSMA ligand-tubulysin compound I to dose level 6, PSMA ligand-tubuly compound I may continue to be dose escalated in 25% increments.
  • PSMA ligand-tubulysin compound I Dose Escalation Scheme: QW schedule cohorts consisting of 3 to 6 patients per dose level will be treated with study therapy following a 3+3 schema. All patients in a dose cohort must complete the Cycle 1 dose-limiting toxicity (DLT) evaluation before dosing is initiated at the next higher dose level. DLTs observed in Cycle 1 will be used to determine whether additional patients should be enrolled at the same dose level, at a lower dose level, or a higher dose level according to the rules outlined below.
  • DLT dose-limiting toxicity
  • DLTs will be based on events occurring during the first cycle of therapy and the adverse events (AEs) must be drug related (i.e. definitely, probably or possibly):
  • Grade 3 neutropenia with fever >38.5°C regardless of use of antibiotic or anti-fungal treatment. • Grade 3 neutropenia persisting for > 5 days.
  • Grade 3 non-hematological toxicity • > Grade 3 non-hematological toxicity.
  • Grade 3 laboratory abnormalities e.g., potassium (K) or Magnesium (Mg)
  • K potassium
  • Mg Magnesium
  • Vllb Schedule Specific DLT's:
  • Schedule #1 TIW Dosing Schedule: Inability to administer at least 4 of the 6 scheduled doses of PSMA ligand-tubulysin compound I in a cycle due to drug-related toxicity.
  • Schedule #2 Once Weekly Dosing Schedule: Inability to administer both scheduled doses of PSMA ligand-tubulysin compound I in a cycle due to drug-related toxicity.
  • intra-patient dose escalation of single agent EC 1169 may be offered starting with Cycle 3 for patients who meet all of the following criteria:
  • PSMA-imaging conjugate Ilia 99m Tc-EC0652
  • Part A Schedule #1: 5 pts; Schedule #2: 35 pts; Part B: Taxane-narve: 14 pts; Taxane-exposed: 18 pts).
  • Anti-tumor activity is suggested with lesion-level reductions.
  • One patient in Part A QW, 3.8 mg/m cohort
  • one patient in Part B QW, 6.5 mg/m taxane-exposed cohort
  • Eight Part A patients have had stable disease lasting at least 3 months, with 3 of those patients having stable disease lasting > 6 months.
  • Eight Part B patients have had stable disease lasting at least 3 months. Both Compound I and PSMA-imaging conjugate Ilia ( 99m Tc-EC0652) were well tolerated in mCRPC patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention décrite dans la présente description concerne des conjugués d'administration de médicaments pour une thérapie ciblée. L'invention décrite dans la présente description concerne des méthodes de traitement de cancers exprimant le PSMA avec un composé ligand de PSMA-tubulysine. L'invention décrite dans la présente description concerne également des méthodes de traitement de cancers exprimant le PSMA avec un composé ligand de PSMA-tubulysine chez des patients, des résultats indiquant une stabilisation de la maladie étant obtenus après traitement avec le composé ligand de PSMA-tubulysine.
PCT/US2017/034130 2016-05-24 2017-05-24 Méthodes de traitement du cancer avec un composé ligand de psma-tubulysine WO2017205447A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662340887P 2016-05-24 2016-05-24
US62/340,887 2016-05-24
US201662345412P 2016-06-03 2016-06-03
US62/345,412 2016-06-03

Publications (1)

Publication Number Publication Date
WO2017205447A1 true WO2017205447A1 (fr) 2017-11-30

Family

ID=60412543

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/034130 WO2017205447A1 (fr) 2016-05-24 2017-05-24 Méthodes de traitement du cancer avec un composé ligand de psma-tubulysine

Country Status (1)

Country Link
WO (1) WO2017205447A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006096754A2 (fr) * 2005-03-07 2006-09-14 Archemix Corp. Aptameres stabilises d'apsm et leur utilisation comme agents therapeutiques contre le cancer de la prostate
US20110172254A1 (en) * 2008-09-17 2011-07-14 Endocyte, Inc. Folate receptor binding conjugates of antifolates
WO2011106639A1 (fr) * 2010-02-25 2011-09-01 Purdue Research Foundation Conjugués ligand de liaison à un antigène de membrane spécifique à la prostate (psma) lieur et procédés d'utilisation associés
WO2014078484A1 (fr) * 2012-11-15 2014-05-22 Endocyte, Inc. Conjugués pour traiter les maladies provoquées par des cellules exprimant psma
US20140154702A1 (en) * 2012-11-30 2014-06-05 Endocyte, Inc. Methods For Treating Cancer Using Combination Therapies

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006096754A2 (fr) * 2005-03-07 2006-09-14 Archemix Corp. Aptameres stabilises d'apsm et leur utilisation comme agents therapeutiques contre le cancer de la prostate
US20110172254A1 (en) * 2008-09-17 2011-07-14 Endocyte, Inc. Folate receptor binding conjugates of antifolates
WO2011106639A1 (fr) * 2010-02-25 2011-09-01 Purdue Research Foundation Conjugués ligand de liaison à un antigène de membrane spécifique à la prostate (psma) lieur et procédés d'utilisation associés
WO2014078484A1 (fr) * 2012-11-15 2014-05-22 Endocyte, Inc. Conjugués pour traiter les maladies provoquées par des cellules exprimant psma
US20150297735A1 (en) * 2012-11-15 2015-10-22 Endocyte, Inc. Conjugates for treating diseases caused by psma expressing cells
US20140154702A1 (en) * 2012-11-30 2014-06-05 Endocyte, Inc. Methods For Treating Cancer Using Combination Therapies

Similar Documents

Publication Publication Date Title
EP3555627B1 (fr) Imagerie et thérapie ciblées par une protéine d'activation des fibroblastes (fap)
JP7184775B2 (ja) 黄体化ホルモン放出ホルモン受容体(lhrh-r)コンジュゲートおよびその使用
US20170290878A1 (en) Methods of treating cancer with tubulysin conjugates
JP2013542263A (ja) 癌を処置する方法
KR20120050462A (ko) 엽산-표적화된 진단법 및 처치법
JP2022527821A (ja) Hsp90結合コンジュゲート及びその製剤
JP2024059640A (ja) 癌を治療する方法
US20180036364A1 (en) Methods of treating cancer with a psma ligand-tubulysin compound
CA3112806A1 (fr) Methodes de traitement de cancer
US20210323985A1 (en) Shielding agents and their use
US20130252904A1 (en) Compositions and methods for treating cancer
WO2017205447A1 (fr) Méthodes de traitement du cancer avec un composé ligand de psma-tubulysine
RU2811406C2 (ru) Способы лечения рака
US20170348376A1 (en) Tubulysin conjugate for use in treating cancer
TW202409009A (zh) 黑素皮質素2型受體(mc2r)之靶向治療劑及其用途
KR20230171964A (ko) 폴레이트 수용체-표적화된 방사선 요법제 및 이의 용도
JP2024518097A (ja) 刷子縁膜酵素切断可能リンカーを持つ葉酸受容体標的コンジュゲート並びにガンのイメージング及び治療における使用方法

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17803471

Country of ref document: EP

Kind code of ref document: A1