WO2020148744A1 - Combination therapy for treatment of pancreatic cancer - Google Patents

Combination therapy for treatment of pancreatic cancer Download PDF

Info

Publication number
WO2020148744A1
WO2020148744A1 PCT/IL2019/051126 IL2019051126W WO2020148744A1 WO 2020148744 A1 WO2020148744 A1 WO 2020148744A1 IL 2019051126 W IL2019051126 W IL 2019051126W WO 2020148744 A1 WO2020148744 A1 WO 2020148744A1
Authority
WO
WIPO (PCT)
Prior art keywords
effective amount
therapeutically effective
chemotherapy
subject
treatment
Prior art date
Application number
PCT/IL2019/051126
Other languages
French (fr)
Inventor
Gideon Stein
Amnon Peled
Original Assignee
Biolinerx Ltd.
Biokine Therapeutics Ltd.
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 Biolinerx Ltd., Biokine Therapeutics Ltd. filed Critical Biolinerx Ltd.
Publication of WO2020148744A1 publication Critical patent/WO2020148744A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention in some embodiments thereof, relates to the treatment of pancreatic cancer.
  • Pancreatic cancer is chemotherapy-resistant, with an extremely poor prognosis. It is the fourth leading cause of cancer death in the United States; the 5-year survival rate is 6 %.
  • the incidence of pancreatic cancer has increased during the past several decades and in 2014, an estimated 46,420 patients were diagnosed with pancreatic cancer and 39,590 died.
  • Pancreatic cancer is projected to surpass liver, breast, prostate, and colorectal cancers to become the second- leading cause of cancer-related death by 2030. These statistics reflect the dire nature of the disease and lack of effective therapies.
  • the location of the tumor results in few early symptoms and is often diagnosed at a late stage as a result.
  • the absence of effective screening tools, and a limited understanding of risk factors means that patients have advanced or metastatic disease at the time of diagnosis. Given the poor prognosis and the low median survival rates of less than one year for patients with metastatic disease, new treatment options are still needed.
  • BL-8040 (previously named BKT140, SEQ ID NO: 1) is a highly selective CXCR4 antagonist.
  • the investigational drug is a 14-residue, cyclic, synthetic peptide capped with an aromatic ring that binds to CXCR4 with high affinity (IC50 0.54-4.5 nM) and inhibits its function[l].
  • the chemokine CXCL12 (SDF-1 -stromal-derived-factor- 1) and its receptor, CXCR4 play a pivotal role in the trafficking of hematopoietic cells to the bone marrow (BM)[2].
  • BL-8040 exhibits a CXCR4-dependent preferential antitumor effect against malignant cells overexpressing CXCR4[3].
  • the efficacy of BL-8040 and its analogs for blocking CXCR4 in vitro and in vivo has been documented in numerous preclinical studies, including in vitro and in vivo models for small cell lung carcinoma, breast cancer, malignant melanoma, neuroblastoma and pancreatic cancer.
  • BL-8040 also affects the trafficking of immune cells to the tumor microenvironment.
  • BL-8040 may eliminate the immunological barrier and allow the accumulation of immune cells within the tumor microenvironment.
  • BL-8040 The nonclinical development of BL-8040 has encompassed a large number of pharmacodynamic, pharmacokinetic (PK), safety pharmacology, and single and repeated dose toxicity studies.
  • BL-8040 exhibits CXCR4-dependent selective cytotoxicity toward malignant cells both in vivo and in vitro and induces apoptotic cell death in cancer cells[3-6].
  • BL-8040 leads to phosphatidylserine extemalization, decreased mitochondrial membrane potential, caspase activation, subsequent sub-Gl arrest and DNA double-stranded breaks in leukemic and multiple myeloma cells [3]. These effects were shown to be specific; BL-8040 did not affect the viability of human keratinocytes and normal human hematopoietic cells [3].
  • BL-8040 This property of direct apoptotic effects on top of the mobilization capacity, distinguishes BL-8040 from other CXCR4 antagonists agents such as Mozobil/Plerixafor [3].
  • administration of BL-8040 induces the mobilization of NK cells, T cells and B cells from the BM and lymph nodes into the periphery.
  • BL-8040 has demonstrated safety and initial clinical efficacy in several Phase I and II studies [Hidalgo MM, Epelbaum R, Semenisty V, Geva R, Golan T, Borazanci EH. Evaluation of pharmacodynamic (PD) biomarkers in patients with metastatic pancreatic cancer treated with BL- 8040, a novel CXCR4 antagonist. J Clin Oncol. 2018; 36:88-88. Abstract].
  • a method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.
  • the anti PD-1 is pembrolizumab.
  • the chemotherapy comprises a single chemotherapeutic agent.
  • the chemotherapy comprises a plurality of chemotherapeutic agents.
  • the chemotherapy comprises irinotecan, fluorouracil (5-FU) and leucovorin (LV).
  • the irinotecan is liposome-encapsulated.
  • the irinotecan is Onivyde®.
  • the chemotherapy comprises gemcitabine.
  • the chemotherapy is selected from the group consisting of gemcitabine, erlotinib, capecitabine, cisplatin and nab-paclitaxel.
  • the peptide is administered subcutaneously (SC) or intravenously (IV).
  • the peptide is administered at a dose of 1.25 mg/kg.
  • the chemotherapy is administered intravenously.
  • the peptide is administered with the chemotherapy.
  • the method or use comprises the use of an anti-histamine and optionally analgesics.
  • the subject is post first-line treatment against the metastatic pancreatic adenocarcinoma.
  • the first-line treatment comprises a gemcitabine-based chemotherapy.
  • the metastatic pancreatic adenocarcinoma is unresectable.
  • the metastatic pancreatic cancer is pancreatic ductal adenocarcinoma.
  • the metastatic pancreatic ductal adenocarcinoma comprises intraductal papillary mucinous neoplasm.
  • the present invention in some embodiments thereof, relates to the treatment of pancreatic cancer.
  • a method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.
  • a chemotherapy for use in treating metastatic pancreatic adenocarcinoma in a subject in need thereof, wherein the subject is not subjected to treatment with an anti-PD- 1.
  • PD- 1 can be present in a first line treatment when the present regimen is used as a second or third line treatment.
  • pancreatic adenocarcinoma refers to stage lib to IV of the disease, when the tumor is present out of the pancreas i.e., lymph nodes or other distal locations.
  • the metastatic pancreatic adenocarcinoma is pancreatic ductal adenocarcinoma.
  • pancreatic ductal adenocarcinoma is a type of exocrine pancreatic cancer. It develops from cells lining small tubes in the pancreas called ducts (duct cells in the diagram above). These carry the digestive juices, which contain enzymes, into the main pancreatic duct and then on into the duodenum (first part of the small intestine). PDAC can grow anywhere in the pancreas, although it is most often found in the head of the pancreas.
  • the PDAC comprises intraductal papillary mucinous neoplasm.
  • the pancreatic cancer is recurrent pancreatic cancer.
  • the pancreatic cancer has reoccurred after remission.
  • the individual has measurable disease (for example, according to RECIST criteria).
  • the individual has one or more metastatic tumors measurable, for example, by CT scan (or MRI).
  • the pancreatic cancer is unresectable pancreatic cancer. In some embodiments, the pancreatic cancer is a resectable pancreatic cancer.
  • the pancreatic cancer is borderline resectable. In some embodiments, the primary location of the pancreatic cancer is the head of the pancreas. In some embodiments, the primary location of the pancreatic cancer is the body of the pancreas. In some embodiments, the primary location of the pancreatic cancer is the tail of the pancreas.
  • subject refers to a human subject diagnosed with metastatic pancreatic adenocarcinoma.
  • the subject is a female.
  • the individual is a male.
  • the individual is under about 65 years old (such as under about any of 60, 55, 50, 45, or 40 years old).
  • the subject is at least about 65 years old (such as at least about any of 70, 75, or 80 years old).
  • the subject is at least 18 years
  • treatment is first-line treatment.
  • treatment is first-line treatment against the pancreatic adenocarcinoma.
  • the treatment is post first-line treatment against the pancreatic adenocarcinoma.
  • methods, regimens, uses, described herein may also be used as a second line or third line therapy after the prior treatment for pancreatic cancer has failed or has substantially failed, or the pancreatic cancer is substantially refractory to the first line therapy.
  • the individual has received at least one line of therapy (e.g., chemotherapy or immunotherapy) for treating metastatic pancreatic cancer prior to receiving the treatment described herein.
  • the patient has received 1 line of therapy or 2 lines of therapy (e.g., 1 line of chemotherapy or immunotherapy).
  • the treatment described herein may be used as a second line therapy.
  • the prior line of therapy described herein may be a prior line of chemotherapy or immunotherapy.
  • the term“treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical symptoms of the metastatic pancreatic adenocarcinoma.
  • the subject is diagnosed with metastatic pancreatic adenocarcinoma.
  • the metastatic pancreatic adenocarcinoma is histologically confirmed (either previously or newly biopsied).
  • the subject has a measurable disease (> 1 measurable lesion) based on Response Evaluation Criteria In Solid Tumors (RECIST) vl.l.
  • a measurable disease > 1 measurable lesion
  • RECIST Response Evaluation Criteria In Solid Tumors
  • tumor lesions situated in a previously irradiated area are considered measurable if progression has been demonstrated in such lesions.
  • the subject has histologically confirmed (either previously or newly biopsied) metastatic unresectable pancreatic adenocarcinoma, including with intraductal papillary mucinous neoplasm.
  • the pancreatic cancer is not acinar cell carcinoma, pancreaticoblastoma, malignant cystic neoplasms, endocrine neoplasms, squamous cell carcinoma.
  • the subject is not immune-deficient.
  • the subject does not have an active autoimmune disease that has required systemic treatment in the 2 years preceding the treatment (i.e., with the use of disease-modifying agents, corticosteroids or immunosuppressive drugs).
  • the subject does not have a history of (non-inf ectious) pneumonitis that required steroids or current pneumonitis.
  • the subject does not have a history of interstitial lung disease.
  • a peptide set forth in SEQ ID NO: 1 also referred to as BL-8040 (previously named“BKT140”) or“the peptide” is a highly selective CXCR4 antagonist, a novel therapy for the treatment of cancer.
  • the peptide is a 14-residue, cyclic, synthetic peptide capped with an aromatic ring that binds to CXCR4 with high affinity (IC50 0.54-4.5 nM) and inhibits its function [Tamamura H, Hiramatsu K, Kusano S, Terakubo S, Yamamoto N, Trent JO, et al. Synthesis of potent CXCR4 inhibitors possessing low cytotoxicity and improved biostability based on T140 derivatives. Org Biomol Chem 2003;1:3656-3662].
  • the BL-8040 is manufactured as a white to off-white powder synthetic polypeptide, freely soluble in water and in 0.45% Sodium Chloride (half normal saline). It is manufactured in accordance with current Good Manufacturing Practice (cGMP) by BioConnection B.V. (previously MSD), Kloosterstraat 9, 5349 AB Oss, Netherlands.
  • cGMP Good Manufacturing Practice
  • the present teachings contemplate the use of chemotherapy for the treatment of the disease.
  • chemotherapy include but are not limited to, gemcitabine, FOLFIRINOX (i.e., oxaliplatin, irinotecan, 5-FU), erlotinib, 5-fluorouracil, paclitaxel, nab-paclitaxel (e.g., Abraxane®), docetaxel, capecitabine, oxaliplatin cisplatin, FOFFOXIRI, an anti-CD40 antibody, oregovomab, Nelfinavir, cetuximab, tegafur, leucovorin, irinotecan and combinations thereof.
  • FOLFIRINOX i.e., oxaliplatin, irinotecan, 5-FU
  • erlotinib e.e., 5-fluorouracil
  • paclitaxel e.g., Abraxane®
  • the chemotherapy is irinotecan, a topoisomerase inhibitor.
  • Irinotecan is converted by esterase enzymes into the more active metabolite, SN-38.
  • the chemical name of irinotecan is (S)-4,l l-diethyl-3,4,12,14-tetrahydro-4-hydroxy-3,14-dioxolH- pyrano[4',4'-:6,7]-indolizino[l,2-b]quinolin-9-yl-[l,4'bipiperidine]-l'-carboxylate.
  • Irinotecan hydrochloride trihydrate is also referred to by the name CPT-11 and by the trade name CAMPTOSAR®.
  • the topoisomerase inhibitor can be camptothecin conjugated to a biocompatible polymer such as a cyclodextrin or cyclodextrin analog (e.g., sulfonated cyclodextrins).
  • the topoisomerase inhibitor can be a cyclodextrin-containing polymer chemically bound to a camptothecin, irinotecan, SN-38 or other topoisomerase 1 inhibitor compound.
  • a cyclodextrin- camptothecin conjugated topoisomerase 1 inhibitor can be administered at a pharmaceutically acceptable dose including 6, 12, or 18 mg/m2 weekly administration, or 12, 15 or 18 mg/m2 biweekly administration.
  • camptothecin-cyclodextrin conjugate topoisomerase 1 inhibitors e.g., the cyclodextrin-containing polymer conjugate with camptothecin designated "CRFX101"
  • camptothecin-cyclodextrin conjugate topoisomerase 1 inhibitors e.g., the cyclodextrin-containing polymer conjugate with camptothecin designated "CRFX101”
  • the topoisomerase inhibitor can also be a liposomal formulation of a topoisomerase inhibitor such as irinotecan, camptothecin or topotecan.
  • a topoisomerase inhibitor such as irinotecan, camptothecin or topotecan.
  • Fiposomal irinotecan e.g., MM-398, also called "nal-IRI”
  • MM-398 also called "nal-IRI”
  • irinotecan is a highly stabilized liposomal formulation of irinotecan that provides for sustained exposure of irinotecan, and the active metabolite SN-38 in the tumor to a higher proportion of cells during the more sensitive S-phase of the cell cycle.
  • MM-398 is a liposomal irinotecan that has shown promising preclinical and clinical activity in a range of cancer types, and was recently approved in the United States in combination with 5-FU/FV for patients with metastatic adenocarcinoma of the pancreas after disease progression following gemcitabine-based therapy.
  • nal-IRI has an extended PK profile with prolonged local tumor exposure of MM-398 and SN-38. Since SN-38 is cleared more quickly from normal tissues than from tumor, it is hypothesized that delayed dosing of veliparib relative to MM-398 will allow for the expected window of maximum irinotecan-induced toxicity to pass in the absence of concurrent veliparib toxicity.
  • the tumor levels of SN-38 are predicted to be sustained upon subsequent veliparib dosing, therefore maintaining the ability of both drugs to act on tumor tissue simultaneously and maintain synergy.
  • liposomal irinotecan available under the brand name ONIVYDE®. (irinotecan liposome injection) (Merrimack Pharmaceuticals, Inc. Cambridge, Mass.), previously designated “MM-398" prior to FDA approval, and liposomal irinotecan products that are bioequivalent to ONIVYDE.
  • the ONIVYDE/MM-398 irinotecan liposome injection
  • the drug product liposome is a small unilamellar lipid bilayer vesicle, approximately 110 nm in diameter, which encapsulates an aqueous space which contains irinotecan in a gelated or precipitated state, as the sucrosofate salt.
  • the liposome carriers are composed of l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 6.81 mg/mL; cholesterol, 2.22 mg/mL; and methoxy-terminated polyethylene glycol (MW 2000)- distearoylphosphatidylethanolamine (MPEG-2000-DSPE), 0.12 mg/mL.
  • Each mL also contains 2-[4-(2-hydroxyethyl)piperazin-l-yl]ethanesulfonic acid (HEPES) as a buffer, 4.05 mg/mL; sodium chloride as isotonicity reagent, 8.42 mg/mL, ONIVYDE/MM-39& is believed to include about 80,000 molecules of irinotecan in a gelated or precipitated state as a sucrosofate salt, encapsulated in a liposome of about 100 nm in diameter.
  • HEPES 2-[4-(2-hydroxyethyl)piperazin-l-yl]ethanesulfonic acid
  • Gemcitabine e.g., GemzarTM
  • Gemcitabine-based chemotherapy e.g., combined with other drugs such as albumin-bound paclitaxel (Abraxane), erlotinib (Tarceva), or capecitabine (Xeloda) or combined with radiation (this is called chemoradiation).
  • other drugs such as albumin-bound paclitaxel (Abraxane), erlotinib (Tarceva), or capecitabine (Xeloda) or combined with radiation (this is called chemoradiation).
  • leucovorin refers to folinic acid that is typically administered in combination with Onivyde(R) and 5-FU.
  • 5-FU is a thymidylate synthase (TS) inhibitor. Interrupting the action of this enzyme blocks synthesis of the pyrimidine thymidine, which is a nucleoside required for DNA replication. Thymidylate synthase methylates deoxyuridine monophosphate (dUMP) to form thymidine monophosphate (dTMP). Administration of 5-FU causes a scarcity in dTMP, so rapidly dividing cancerous cells undergo cell death via thymineless death. Calcium folinate provides an exogenous source of reduced folinates and hence stabilizes the 5-FU-TS complex, hence enhancing 5-FU's cytotoxicity. 5-FU is sold under the brand name Adrucil, among others.
  • the chemotherapy comprises a single chemotherapeutic agent.
  • the chemotherapy comprises a plurality of chemotherapeutic agents e.g., at least 2, or at least 3, e.g., 2, 3, 4, 5 chemotherapeutic agents).
  • the chemotherapy comprises irinotecan (e.g., liposome-encapsulated e.g., Onivyde (R) ), 5-FU and Leucovorin.
  • irinotecan e.g., liposome-encapsulated e.g., Onivyde (R)
  • 5-FU e.g., 5-FU
  • Leucovorin e.g., 5-FU
  • the chemotherapy comprises gemcitabine, fixed dose rate gemcitabine, gemcitabine with cisplatin, gemcitabine with capecitabine, gemcitabine with erlotinib or gemcitabine with nab-paclitaxel.
  • the chemotherapy comprises, fixed dose rate gemcitabine (GTX) with docetaxel and capecitabine, 5-Fu and leucovirin or capecitabine and oxaliplatin.
  • GTX fixed dose rate gemcitabine
  • treatment does not include oxaliplatin.
  • agents can be administered to the subject per se, or in a pharmaceutical composition being mixed with suitable carriers or excipients.
  • agents can be formulated in a separate formulation or at least some of them combined to a single formulation.
  • a "pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • active ingredient refers to the agents accountable for the biological effect, e.g., SEQ ID NO: 1 and chemotherapy.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, intradermal, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • the peptide of the invention or the pharmaceutical composition comprising same is administered subcutaneously (SC).
  • the peptide of the invention or the pharmaceutical composition comprising same is administered intravenously (IV).
  • the chemotherapy or the pharmaceutical composition comprising same is administered intravenously.
  • compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with some embodiments of the invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, according to specific embodiments, a therapeutically effective amount means an amount of active ingredients effective to prevent, alleviate or ameliorate symptoms of a disorder (i.e., metastatic pancreatic adenocarcinoma) or prolong the survival of the subject being treated. According to specific embodiments the peptide of the invention or the pharmaceutical composition comprising same is administered in a dose ranging between 0.1 to 10 mg/kg of body weight, between 0.1 to 2 mg/kg of body weight, between 0.1 to 1 mg/kg of body weight, between 0.3 to 10 mg/kg of body weight, between 0.3 to 2.
  • a therapeutically effective amount means an amount of active ingredients effective to prevent, alleviate or ameliorate symptoms of a disorder (i.e., metastatic pancreatic adenocarcinoma) or prolong the survival of the subject being treated.
  • the peptide of the invention or the pharmaceutical composition comprising same is
  • BL-8040 is administered at a dose of 1-2 mg/kg body weight.
  • the, BL-8040 is administered at a dose of 1.25-1.5 mg/kg body weight.
  • the BL-8040 is administered at a dose of 1.25 mg/kg body weight.
  • the BL-8040 is administered subcutaneously (SC).
  • the chemotherapy is administered intravenously.
  • the peptide is administered with the chemotherapy (e.g., on the same day) or at least one of the chemotherapies.
  • the peptide is administered in the same formulation as at least one of the chemotherapies, or in separate formulations.
  • the chemotherapy is administered according to the tolerated dose which is known in the field.
  • Gemcitabine e.g., 1,000 mg/m2
  • Erlotinib is administered daily (e.g., 100 mg per day) throughout the treatment period.
  • the peptide is administered IV or SC with each gemcitabine dose.
  • the peptide is administered IV or SC with each gemcitabine dose.
  • the peptide is administered IV or SC with each gemcitabine dose.
  • Gemcitabine e.g., 1000 mg/m 2 IV
  • nab-paclitaxel e.g., 125 mg/m 2 IV
  • the peptide is administered IV or SC with each treatment.
  • Onivyde is administered prior to LV and 5-FU.
  • Patients homozygous for the UGT1A1*28 allele will initiate Onivyde ® at 50 mg/m 2 , and the dose can be increased if tolerated at later (e.g., 35 cycles).
  • the peptide is administered IV or SC with each irinotecan liposome dose once every two weeks.
  • Efficacy testing can be done at any time, e.g., such as prior to, during or after the execution of the regimen as described herein e.g., following at least 1, 2, 3, 5, 10 or more cycles of the therapy.
  • imaging is used for assessment of response.
  • CT or MRI may be used.
  • the same imaging method may be preferably used throughout the treatment for each subject.
  • a biopsy can be performed using known tools available for those of skills in the art of pathology.
  • samples are collected for safety and efficacy analysis, e.g., CBC, anti-drug antibodies titer and determination of BL-8040 plasma concentrations.
  • compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed above.
  • compositions, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • a compound or “at least one compound” may include a plurality of compounds, including mixtures thereof.
  • range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • method refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.

Abstract

A method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof is provided. The method comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.

Description

COMBINATION THERAPY FOR TREATMENT OF PANCREATIC CANCER
REPLATED APPLICATION
This application claims the benefit of priority from U.S. Provisional Patent Application No. 62/793,410, filed 17 January 2019, which is hereby incorporated in its entirety.
SEQUENCE LISTING STATEMENT
The ASCII file, entitled 78929 Sequence Listing.txt, created on 8 September 2019, comprising 4,096 bytes, submitted concurrently with the filing of this application is incorporated herein by reference.
FIELD AND BACKGROUND OF THE INVENTION
The present invention, in some embodiments thereof, relates to the treatment of pancreatic cancer.
Pancreatic cancer is chemotherapy-resistant, with an extremely poor prognosis. It is the fourth leading cause of cancer death in the United States; the 5-year survival rate is 6 %. The incidence of pancreatic cancer has increased during the past several decades and in 2014, an estimated 46,420 patients were diagnosed with pancreatic cancer and 39,590 died. Pancreatic cancer is projected to surpass liver, breast, prostate, and colorectal cancers to become the second- leading cause of cancer-related death by 2030. These statistics reflect the dire nature of the disease and lack of effective therapies. The location of the tumor results in few early symptoms and is often diagnosed at a late stage as a result. The absence of effective screening tools, and a limited understanding of risk factors, means that patients have advanced or metastatic disease at the time of diagnosis. Given the poor prognosis and the low median survival rates of less than one year for patients with metastatic disease, new treatment options are still needed.
BL-8040 (previously named BKT140, SEQ ID NO: 1) is a highly selective CXCR4 antagonist. The investigational drug is a 14-residue, cyclic, synthetic peptide capped with an aromatic ring that binds to CXCR4 with high affinity (IC50 0.54-4.5 nM) and inhibits its function[l]. The chemokine CXCL12 (SDF-1 -stromal-derived-factor- 1) and its receptor, CXCR4, play a pivotal role in the trafficking of hematopoietic cells to the bone marrow (BM)[2]. Animal studies exploring the activity of BL-8040 in various cancer models in mice have shown that in addition to its activity as a mobilizer of hematopoietic cells, BL-8040 exhibits a CXCR4- dependent preferential antitumor effect against malignant cells overexpressing CXCR4[3]. The efficacy of BL-8040 and its analogs for blocking CXCR4 in vitro and in vivo has been documented in numerous preclinical studies, including in vitro and in vivo models for small cell lung carcinoma, breast cancer, malignant melanoma, neuroblastoma and pancreatic cancer. As a CXCR4 antagonist, BL-8040 also affects the trafficking of immune cells to the tumor microenvironment. It was found that administration of BL-8040 induces the mobilization of Natural Killer (NK) cells, T cells and B cells from the BM and lymph nodes into the periphery. Using a syngeneic cancer model in mice it was demonstrated that BL-8040 may eliminate the immunological barrier and allow the accumulation of immune cells within the tumor microenvironment.
Preclinical Studies
The nonclinical development of BL-8040 has encompassed a large number of pharmacodynamic, pharmacokinetic (PK), safety pharmacology, and single and repeated dose toxicity studies.
BL-8040 exhibits CXCR4-dependent selective cytotoxicity toward malignant cells both in vivo and in vitro and induces apoptotic cell death in cancer cells[3-6]. BL-8040 leads to phosphatidylserine extemalization, decreased mitochondrial membrane potential, caspase activation, subsequent sub-Gl arrest and DNA double-stranded breaks in leukemic and multiple myeloma cells [3]. These effects were shown to be specific; BL-8040 did not affect the viability of human keratinocytes and normal human hematopoietic cells [3]. This property of direct apoptotic effects on top of the mobilization capacity, distinguishes BL-8040 from other CXCR4 antagonists agents such as Mozobil/Plerixafor [3]. In addition, administration of BL-8040 induces the mobilization of NK cells, T cells and B cells from the BM and lymph nodes into the periphery.
BL-8040 has demonstrated safety and initial clinical efficacy in several Phase I and II studies [Hidalgo MM, Epelbaum R, Semenisty V, Geva R, Golan T, Borazanci EH. Evaluation of pharmacodynamic (PD) biomarkers in patients with metastatic pancreatic cancer treated with BL- 8040, a novel CXCR4 antagonist. J Clin Oncol. 2018; 36:88-88. Abstract].
Additional background art includes:
W02017009843
W02017009842.
SUMMARY OF THE INVENTION
According to an aspect of some embodiments of the present invention there is provided a method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof, the method comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.
According to an aspect of some embodiments of the present invention there is provided a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy for use in treating metastatic pancreatic adenocarcinoma in a subject in need thereof, wherein the subject is not subjected to treatment with an anti-PD-1.
According to some embodiments of the invention, the anti PD-1 is pembrolizumab.
According to some embodiments of the invention, the chemotherapy comprises a single chemotherapeutic agent.
According to some embodiments of the invention, the chemotherapy comprises a plurality of chemotherapeutic agents.
According to some embodiments of the invention, the chemotherapy comprises irinotecan, fluorouracil (5-FU) and leucovorin (LV).
According to some embodiments of the invention, the irinotecan is liposome-encapsulated.
According to some embodiments of the invention, the irinotecan is Onivyde®.
According to some embodiments of the invention, the chemotherapy comprises gemcitabine.
According to some embodiments of the invention, the chemotherapy is selected from the group consisting of gemcitabine, erlotinib, capecitabine, cisplatin and nab-paclitaxel.
According to some embodiments of the invention, the peptide is administered subcutaneously (SC) or intravenously (IV).
According to some embodiments of the invention, the peptide is administered at a dose of 1.25 mg/kg.
According to some embodiments of the invention, the chemotherapy is administered intravenously.
According to some embodiments of the invention, the peptide is administered with the chemotherapy.
According to some embodiments of the invention, the method or use comprises the use of an anti-histamine and optionally analgesics.
According to some embodiments of the invention, the subject is post first-line treatment against the metastatic pancreatic adenocarcinoma. According to some embodiments of the invention, the first-line treatment comprises a gemcitabine-based chemotherapy.
According to some embodiments of the invention, the metastatic pancreatic adenocarcinoma is unresectable.
According to some embodiments of the invention, the metastatic pancreatic cancer is pancreatic ductal adenocarcinoma.
According to some embodiments of the invention, the metastatic pancreatic ductal adenocarcinoma comprises intraductal papillary mucinous neoplasm.
Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.
DESCRIPTION OF SPECIFIC EMBODIMENTS OF THE INVENTION
The present invention, in some embodiments thereof, relates to the treatment of pancreatic cancer.
Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.
According to an aspect of the invention there is provided a method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof, the method comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.
According to an aspect of the invention there is provided a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy for use in treating metastatic pancreatic adenocarcinoma in a subject in need thereof, wherein the subject is not subjected to treatment with an anti-PD- 1. As used herein“not subjected to treatment with an anti-PD-1” means in the same cycle of treatment. PD- 1 can be present in a first line treatment when the present regimen is used as a second or third line treatment.
As used herein“metastatic pancreatic adenocarcinoma” refers to stage lib to IV of the disease, when the tumor is present out of the pancreas i.e., lymph nodes or other distal locations.
Table 1-Stages of pancreatic cancer
Figure imgf000006_0001
Figure imgf000007_0001
Adapted from - /www(dot)cancer(dot)org/cancer/pancreatic-cancer/detection-diagnosis-staging/staging(dot)html
According to a specific embodiment, the metastatic pancreatic adenocarcinoma is pancreatic ductal adenocarcinoma.
As used herein “pancreatic ductal adenocarcinoma” (PDAC) is a type of exocrine pancreatic cancer. It develops from cells lining small tubes in the pancreas called ducts (duct cells in the diagram above). These carry the digestive juices, which contain enzymes, into the main pancreatic duct and then on into the duodenum (first part of the small intestine). PDAC can grow anywhere in the pancreas, although it is most often found in the head of the pancreas.
According to a specific embodiment, the PDAC comprises intraductal papillary mucinous neoplasm.
In some embodiments, the pancreatic cancer is recurrent pancreatic cancer.
In some embodiments, the pancreatic cancer has reoccurred after remission.
In some embodiments, the individual has measurable disease (for example, according to RECIST criteria).
In some embodiments, the individual has one or more metastatic tumors measurable, for example, by CT scan (or MRI).
In some embodiments, the pancreatic cancer is unresectable pancreatic cancer. In some embodiments, the pancreatic cancer is a resectable pancreatic cancer.
In some embodiments, the pancreatic cancer is borderline resectable. In some embodiments, the primary location of the pancreatic cancer is the head of the pancreas. In some embodiments, the primary location of the pancreatic cancer is the body of the pancreas. In some embodiments, the primary location of the pancreatic cancer is the tail of the pancreas.
As used herein“subject” refers to a human subject diagnosed with metastatic pancreatic adenocarcinoma.
In some embodiments, the subject is a female.
In some embodiments, the individual is a male.
In some embodiments, the individual is under about 65 years old (such as under about any of 60, 55, 50, 45, or 40 years old).
In some embodiments, the subject is at least about 65 years old (such as at least about any of 70, 75, or 80 years old).
According to a specific embodiment, the subject is at least 18 years
According to a specific embodiment, the subject has not been treated with any previous systemic chemotherapy for the pancreatic adenocarcinoma. Accordingly, treatment is first-line treatment.
According to another embodiment, treatment is first-line treatment against the pancreatic adenocarcinoma.
According to a specific embodiment, the treatment is post first-line treatment against the pancreatic adenocarcinoma.
Thus, methods, regimens, uses, described herein may also be used as a second line or third line therapy after the prior treatment for pancreatic cancer has failed or has substantially failed, or the pancreatic cancer is substantially refractory to the first line therapy. In some embodiments, the individual has received at least one line of therapy (e.g., chemotherapy or immunotherapy) for treating metastatic pancreatic cancer prior to receiving the treatment described herein. In some embodiments, the patient has received 1 line of therapy or 2 lines of therapy (e.g., 1 line of chemotherapy or immunotherapy). Thus, the treatment described herein may be used as a second line therapy. The prior line of therapy described herein may be a prior line of chemotherapy or immunotherapy.
As used herein, the term“treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical symptoms of the metastatic pancreatic adenocarcinoma. According to a specific embodiment, the subject is diagnosed with metastatic pancreatic adenocarcinoma.
According to a specific embodiment the metastatic pancreatic adenocarcinoma is histologically confirmed (either previously or newly biopsied).
According to a specific embodiment, the subject has a measurable disease (> 1 measurable lesion) based on Response Evaluation Criteria In Solid Tumors (RECIST) vl.l.
According to a specific embodiment, tumor lesions situated in a previously irradiated area are considered measurable if progression has been demonstrated in such lesions.
According to a specific embodiment, the subject has histologically confirmed (either previously or newly biopsied) metastatic unresectable pancreatic adenocarcinoma, including with intraductal papillary mucinous neoplasm.
According to a specific embodiment the pancreatic cancer is not acinar cell carcinoma, pancreaticoblastoma, malignant cystic neoplasms, endocrine neoplasms, squamous cell carcinoma. Vater and periampullary duodenal or common bile duct malignancies.
According to a specific embodiment the subject does not have a bowel obstruction
According to a specific embodiment, the subject is not immune-deficient.
According to a specific embodiment, the subject does not have an active autoimmune disease that has required systemic treatment in the 2 years preceding the treatment (i.e., with the use of disease-modifying agents, corticosteroids or immunosuppressive drugs).
According to a specific embodiment, the subject does not have a history of (non-inf ectious) pneumonitis that required steroids or current pneumonitis.
According to a specific embodiment, the subject does not have a history of interstitial lung disease.
As used herein“a peptide set forth in SEQ ID NO: 1” also referred to as BL-8040 (previously named“BKT140”) or“the peptide” is a highly selective CXCR4 antagonist, a novel therapy for the treatment of cancer. The peptide is a 14-residue, cyclic, synthetic peptide capped with an aromatic ring that binds to CXCR4 with high affinity (IC50 0.54-4.5 nM) and inhibits its function [Tamamura H, Hiramatsu K, Kusano S, Terakubo S, Yamamoto N, Trent JO, et al. Synthesis of potent CXCR4 inhibitors possessing low cytotoxicity and improved biostability based on T140 derivatives. Org Biomol Chem 2003;1:3656-3662].
According to a specific embodiment, the BL-8040 is manufactured as a white to off-white powder synthetic polypeptide, freely soluble in water and in 0.45% Sodium Chloride (half normal saline). It is manufactured in accordance with current Good Manufacturing Practice (cGMP) by BioConnection B.V. (previously MSD), Kloosterstraat 9, 5349 AB Oss, Netherlands.
As mentioned, the present teachings contemplate the use of chemotherapy for the treatment of the disease. Examples include but are not limited to, gemcitabine, FOLFIRINOX (i.e., oxaliplatin, irinotecan, 5-FU), erlotinib, 5-fluorouracil, paclitaxel, nab-paclitaxel (e.g., Abraxane®), docetaxel, capecitabine, oxaliplatin cisplatin, FOFFOXIRI, an anti-CD40 antibody, oregovomab, Nelfinavir, cetuximab, tegafur, leucovorin, irinotecan and combinations thereof.
According to a specific embodiment the chemotherapy is irinotecan, a topoisomerase inhibitor.
Irinotecan is converted by esterase enzymes into the more active metabolite, SN-38. The chemical name of irinotecan is (S)-4,l l-diethyl-3,4,12,14-tetrahydro-4-hydroxy-3,14-dioxolH- pyrano[4',4'-:6,7]-indolizino[l,2-b]quinolin-9-yl-[l,4'bipiperidine]-l'-carboxylate. Irinotecan hydrochloride trihydrate is also referred to by the name CPT-11 and by the trade name CAMPTOSAR®.
The topoisomerase inhibitor can be camptothecin conjugated to a biocompatible polymer such as a cyclodextrin or cyclodextrin analog (e.g., sulfonated cyclodextrins). For example, the topoisomerase inhibitor can be a cyclodextrin-containing polymer chemically bound to a camptothecin, irinotecan, SN-38 or other topoisomerase 1 inhibitor compound. A cyclodextrin- camptothecin conjugated topoisomerase 1 inhibitor can be administered at a pharmaceutically acceptable dose including 6, 12, or 18 mg/m2 weekly administration, or 12, 15 or 18 mg/m2 biweekly administration. Examples of camptothecin-cyclodextrin conjugate topoisomerase 1 inhibitors (e.g., the cyclodextrin-containing polymer conjugate with camptothecin designated "CRFX101"), and related intermediates for preparing the same, are disclosed, for example, in Greenwald et ak, Bioorg. Med. Chem., 1998, 6, 551-562, as well as United States Patent Application 2010/0247668, United States Patent Application 2011/0160159 and United States Patent Application 2011/0189092.
The topoisomerase inhibitor can also be a liposomal formulation of a topoisomerase inhibitor such as irinotecan, camptothecin or topotecan. Fiposomal irinotecan (e.g., MM-398, also called "nal-IRI") is a highly stabilized liposomal formulation of irinotecan that provides for sustained exposure of irinotecan, and the active metabolite SN-38 in the tumor to a higher proportion of cells during the more sensitive S-phase of the cell cycle. MM-398 is a liposomal irinotecan that has shown promising preclinical and clinical activity in a range of cancer types, and was recently approved in the United States in combination with 5-FU/FV for patients with metastatic adenocarcinoma of the pancreas after disease progression following gemcitabine-based therapy. Compared with free irinotecan, nal-IRI has an extended PK profile with prolonged local tumor exposure of MM-398 and SN-38. Since SN-38 is cleared more quickly from normal tissues than from tumor, it is hypothesized that delayed dosing of veliparib relative to MM-398 will allow for the expected window of maximum irinotecan-induced toxicity to pass in the absence of concurrent veliparib toxicity. However, the tumor levels of SN-38 are predicted to be sustained upon subsequent veliparib dosing, therefore maintaining the ability of both drugs to act on tumor tissue simultaneously and maintain synergy.
One suitable liposomal Topi inhibitor formulation is liposomal irinotecan available under the brand name ONIVYDE®. (irinotecan liposome injection) (Merrimack Pharmaceuticals, Inc. Cambridge, Mass.), previously designated "MM-398" prior to FDA approval, and liposomal irinotecan products that are bioequivalent to ONIVYDE. The ONIVYDE/MM-398 (irinotecan liposome injection) includes irinotecan as an irinotecan sucrosofate salt encapsulated in liposomes for intravenous use. The drug product liposome is a small unilamellar lipid bilayer vesicle, approximately 110 nm in diameter, which encapsulates an aqueous space which contains irinotecan in a gelated or precipitated state, as the sucrosofate salt. The liposome carriers are composed of l,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 6.81 mg/mL; cholesterol, 2.22 mg/mL; and methoxy-terminated polyethylene glycol (MW 2000)- distearoylphosphatidylethanolamine (MPEG-2000-DSPE), 0.12 mg/mL. Each mL also contains 2-[4-(2-hydroxyethyl)piperazin-l-yl]ethanesulfonic acid (HEPES) as a buffer, 4.05 mg/mL; sodium chloride as isotonicity reagent, 8.42 mg/mL, ONIVYDE/MM-39& is believed to include about 80,000 molecules of irinotecan in a gelated or precipitated state as a sucrosofate salt, encapsulated in a liposome of about 100 nm in diameter.
Gemcitabine (e.g., Gemzar™) or Gemcitabine-based chemotherapy e.g., combined with other drugs such as albumin-bound paclitaxel (Abraxane), erlotinib (Tarceva), or capecitabine (Xeloda) or combined with radiation (this is called chemoradiation).
As used herein “leucovorin” refers to folinic acid that is typically administered in combination with Onivyde(R) and 5-FU.
5-FU is a thymidylate synthase (TS) inhibitor. Interrupting the action of this enzyme blocks synthesis of the pyrimidine thymidine, which is a nucleoside required for DNA replication. Thymidylate synthase methylates deoxyuridine monophosphate (dUMP) to form thymidine monophosphate (dTMP). Administration of 5-FU causes a scarcity in dTMP, so rapidly dividing cancerous cells undergo cell death via thymineless death. Calcium folinate provides an exogenous source of reduced folinates and hence stabilizes the 5-FU-TS complex, hence enhancing 5-FU's cytotoxicity. 5-FU is sold under the brand name Adrucil, among others.
According to a specific embodiment, the chemotherapy comprises a single chemotherapeutic agent.
According to a specific embodiment, the chemotherapy comprises a plurality of chemotherapeutic agents e.g., at least 2, or at least 3, e.g., 2, 3, 4, 5 chemotherapeutic agents).
According to a specific embodiment, the chemotherapy comprises irinotecan (e.g., liposome-encapsulated e.g., Onivyde(R)), 5-FU and Leucovorin.
According to a specific embodiment, the chemotherapy comprises gemcitabine, fixed dose rate gemcitabine, gemcitabine with cisplatin, gemcitabine with capecitabine, gemcitabine with erlotinib or gemcitabine with nab-paclitaxel.
According to a specific embodiment, the chemotherapy comprises, fixed dose rate gemcitabine (GTX) with docetaxel and capecitabine, 5-Fu and leucovirin or capecitabine and oxaliplatin.
Doses and regimen for chemotherapy treatment of the disease as described herein and according to performance status are well known in the art.
According to a specific embodiment treatment does not include oxaliplatin.
The peptide and chemotherapy (“agents”) described hereinabove can be administered to the subject per se, or in a pharmaceutical composition being mixed with suitable carriers or excipients. Each of the agents can be formulated in a separate formulation or at least some of them combined to a single formulation.
As used herein a "pharmaceutical composition" refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
Herein the term "active ingredient" refers to the agents accountable for the biological effect, e.g., SEQ ID NO: 1 and chemotherapy.
Hereinafter, the phrases "physiologically acceptable carrier" and "pharmaceutically acceptable carrier" which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.
Herein the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
Techniques for formulation and administration of drugs may be found in“Remington’s Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, intradermal, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intraocular injections.
Alternately, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a tissue region of a patient.
According to a specific embodiment, the peptide of the invention or the pharmaceutical composition comprising same is administered subcutaneously (SC).
According to a specific embodiment, the peptide of the invention or the pharmaceutical composition comprising same is administered intravenously (IV).
According to a specific embodiment, the chemotherapy or the pharmaceutical composition comprising same is administered intravenously.
Pharmaceutical compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
Pharmaceutical compositions for use in accordance with some embodiments of the invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, according to specific embodiments, a therapeutically effective amount means an amount of active ingredients effective to prevent, alleviate or ameliorate symptoms of a disorder (i.e., metastatic pancreatic adenocarcinoma) or prolong the survival of the subject being treated. According to specific embodiments the peptide of the invention or the pharmaceutical composition comprising same is administered in a dose ranging between 0.1 to 10 mg/kg of body weight, between 0.1 to 2 mg/kg of body weight, between 0.1 to 1 mg/kg of body weight, between 0.3 to 10 mg/kg of body weight, between 0.3 to 2.
According to a specific embodiment, BL-8040 is administered at a dose of 1-2 mg/kg body weight.
According to a specific embodiment, the, BL-8040 is administered at a dose of 1.25-1.5 mg/kg body weight.
According to a specific embodiment, the BL-8040 is administered at a dose of 1.25 mg/kg body weight.
According to a specific embodiment, the BL-8040 is administered subcutaneously (SC). According to a specific embodiment, the chemotherapy is administered intravenously. According to a specific embodiment, the peptide is administered with the chemotherapy (e.g., on the same day) or at least one of the chemotherapies. According to a specific embodiment, the peptide is administered in the same formulation as at least one of the chemotherapies, or in separate formulations.
According to a specific embodiment, the chemotherapy is administered according to the tolerated dose which is known in the field.
Exemplary therapeutic regimen according to some embodiments of the invention
I. Gemcitabine with erlotinib+BL-8040:
Gemcitabine (e.g., 1,000 mg/m2) is administered weekly for up to 6 weeks followed by a week of rest and then once weekly for 3 consecutive weeks out of every 4 weeks. Erlotinib is administered daily (e.g., 100 mg per day) throughout the treatment period. The peptide is administered IV or SC with each gemcitabine dose.
II. Gemcitabine with capecitabine+BL-8040:
The peptide is administered IV or SC with each gemcitabine dose.
III. Gemcitabine with cisplatin
The peptide is administered IV or SC with each gemcitabine dose.
IV. Gemcitabine with nab-paclitaxel (Abraxane®) + BL-8040:
Gemcitabine (e.g., 1000 mg/m2 IV) and nab-paclitaxel (e.g., 125 mg/m2 IV) are administered once a week on the same day, for 3 weeks and then followed by treatment every 28 days. The peptide is administered IV or SC with each treatment. V. Irinotecan liposome (Onivyde) in combination with fluorouracil and leucovorin+BL-8040:
Onivyde is administered prior to LV and 5-FU. Onivyde 70 mg/m2 as an IV infusion over 90 minutes, followed by LV 400 mg/m2 IV over 30 minutes, followed by 5-FU 2400 mg/m2 IV over 46 hours, every 2 weeks. Patients homozygous for the UGT1A1*28 allele will initiate Onivyde® at 50 mg/m2, and the dose can be increased if tolerated at later (e.g., 35 cycles).
The peptide is administered IV or SC with each irinotecan liposome dose once every two weeks.
Efficacy testing can be done at any time, e.g., such as prior to, during or after the execution of the regimen as described herein e.g., following at least 1, 2, 3, 5, 10 or more cycles of the therapy.
According to a specific embodiment, imagining is used for assessment of response. For example, CT or MRI may be used. The same imaging method may be preferably used throughout the treatment for each subject.
Alternatively or additionally, a biopsy can be performed using known tools available for those of skills in the art of pathology.
Alternatively or additionally, samples are collected for safety and efficacy analysis, e.g., CBC, anti-drug antibodies titer and determination of BL-8040 plasma concentrations.
Compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed above.
It is expected that during the life of a patent maturing from this application many relevant chemotherapies for the treatment of pancreatic cancer will be developed and the scope of the term is intended to include all such new technologies a priori. As used herein the term“about” refers to ± 10 %.
The terms "comprises", "comprising", "includes", "including", “having” and their conjugates mean "including but not limited to".
The term“consisting of’ means“including and limited to”.
The term "consisting essentially of" means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
As used herein, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a compound" or "at least one compound" may include a plurality of compounds, including mixtures thereof.
Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases“ranging/ranges between” a first indicate number and a second indicate number and“ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
As used herein the term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.
In addition, any priority document(s) of this application is/are hereby incorporated herein by reference in its/their entirety.

Claims

WHAT IS CLAIMED IS:
1. A method of treating metastatic pancreatic adenocarcinoma in a subject in need thereof, the method comprising, administering to the subject a therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy, thereby treating the metastatic pancreatic adenocarcinoma, wherein the subject is not subjected to treatment with an anti-PD-1.
2. A therapeutically effective amount of a peptide set forth in SEQ ID NO: 1 and a chemotherapy for use in treating metastatic pancreatic adenocarcinoma in a subject in need thereof, wherein the subject is not subjected to treatment with an anti-PD-1.
3. The method or therapeutically effective amount of any one of claims 1-2, wherein said anti PD-1 is pembrolizumab.
4. The method or therapeutically effective amount of any one of claims 1-3, wherein said chemotherapy comprises a single chemotherapeutic agent.
5. The method or therapeutically effective amount of any one of claims 1-3, wherein said chemotherapy comprises a plurality of chemotherapeutic agents.
6. The method or therapeutically effective amount of any one of claims 1-3 or 5, wherein said chemotherapy comprises irinotecan, fluorouracil (5-FU) and leucovorin (LV).
7. The method or therapeutically effective amount of any one of claims 1-3 or 5-6, wherein said irinotecan is liposome-encapsulated.
8. The method or therapeutically effective amount of any one of claims 1-3 or 5-7, wherein said irinotecan is Onivyde®.
9. The method or therapeutically effective amount of any one of claims 1-5, wherein said chemotherapy comprises gemcitabine.
10. The method or therapeutically effective amount of any one of claims 1-5, wherein said chemotherapy is selected from the group consisting of gemcitabine, erlotinib, capecitabine, cisplatin and nab-paclitaxel.
11. The method or therapeutically effective amount of any one of claims 1-10, wherein said peptide is administered subcutaneously (SC) or intravenously (IV).
12. The method or therapeutically effective amount of any one of claims 1-11, wherein said peptide is administered at a dose of 1.25 mg/kg.
13. The method or therapeutically effective amount of any one of claims 1-12, wherein said chemotherapy is administered intravenously.
14. The method or therapeutically effective amount of any one of claims 1-13, wherein said peptide is administered with said chemotherapy.
15. The method or therapeutically effective amount of any one of claims 1-14, comprising an anti-histamine and optionally analgesics.
16. The method or therapeutically effective amount of any one of claims 1-15, wherein said subject is post first-line treatment against said metastatic pancreatic adenocarcinoma.
17. The method or therapeutically effective amount of claim 16, wherein said first-line treatment comprises a gemcitabine-based chemotherapy.
18. The method or therapeutically effective amount of any one of claims 1-16, wherein said metastatic pancreatic adenocarcinoma is unresectable.
19. The method or therapeutically effective amount of any one of claims 1-16, wherein said metastatic pancreatic cancer is pancreatic ductal adenocarcinoma.
20. The method or therapeutically effective amount of claim 19, wherein said metastatic pancreatic ductal adenocarcinoma comprises intraductal papillary mucinous neoplasm.
PCT/IL2019/051126 2019-01-17 2019-10-17 Combination therapy for treatment of pancreatic cancer WO2020148744A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962793410P 2019-01-17 2019-01-17
US62/793,410 2019-01-17

Publications (1)

Publication Number Publication Date
WO2020148744A1 true WO2020148744A1 (en) 2020-07-23

Family

ID=68387380

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2019/051126 WO2020148744A1 (en) 2019-01-17 2019-10-17 Combination therapy for treatment of pancreatic cancer

Country Status (1)

Country Link
WO (1) WO2020148744A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100247668A1 (en) 2009-03-30 2010-09-30 Scott Eliasof Polymer-agent conjugates, particles, compositions, and related methods of use
US20110160159A1 (en) 2009-09-15 2011-06-30 John Ryan Treatment of cancer
WO2013188586A1 (en) * 2012-06-13 2013-12-19 Merrimack Pharmaceuticals, Inc. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
WO2017034957A1 (en) * 2015-08-21 2017-03-02 Merrimack Pharmaceuticals, Inc. Methods for treating metastatic pancreatic cancer using combination therapies comprising liposomal irinotecan and oxaliplatin
WO2018083470A1 (en) * 2016-11-02 2018-05-11 Ipsen Biopharm Ltd. Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoruracil (and leucovorin)

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100247668A1 (en) 2009-03-30 2010-09-30 Scott Eliasof Polymer-agent conjugates, particles, compositions, and related methods of use
US20110189092A1 (en) 2009-03-30 2011-08-04 Scott Eliasof Polymer-agent conjugates, particles, compositions, and related methods of use
US20110160159A1 (en) 2009-09-15 2011-06-30 John Ryan Treatment of cancer
WO2013188586A1 (en) * 2012-06-13 2013-12-19 Merrimack Pharmaceuticals, Inc. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
WO2017009843A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions, articles of manufacture and methods for treating cancer
WO2017034957A1 (en) * 2015-08-21 2017-03-02 Merrimack Pharmaceuticals, Inc. Methods for treating metastatic pancreatic cancer using combination therapies comprising liposomal irinotecan and oxaliplatin
WO2018083470A1 (en) * 2016-11-02 2018-05-11 Ipsen Biopharm Ltd. Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoruracil (and leucovorin)

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; November 2009 (2009-11-01), BEIDER KATIA ET AL: "Anti-Leukemia and Multiple Myeloma Selective Activity of CXCR4 Antagonist 4F-Benzoyl-TN14003 Involves Apoptotic Death Pathway", XP002796778, Database accession no. PREV201000354849 *
GREENWALD ET AL., BIOORG. MED. CHEM., vol. 6, 1998, pages 551 - 562
HIDALGO MMEPELBAUM RSEMENISTY VGEVA RGOLAN TBORAZANCI EH: "Evaluation of pharmacodynamic (PD) biomarkers in patients with metastatic pancreatic cancer treated with BL-8040, a novel CXCR4 antagonist", J CLIN ONCOL., vol. 36, 2018, pages 88 - 88
MORI T ET AL: "INVOLVEMENT OF STROMAL CELL-DERIVED FACTOR 1 AND CXCR4 RECEPTOR SYSTEM IN PANCREATIC CANCER", GASTROENTEROLOGY : OFFICIAL PUBLICATION OF THE AMERICAN GASTROENTEROLOGICAL ASSOCIATION, WILLIAMS & WILKINS, US, vol. 122, no. 4, Suppl. 1, 1 April 2002 (2002-04-01), pages A490, XP009021758, ISSN: 0016-5085 *
TAMAMURA HHIRAMATSU KKUSANO STERAKUBO SYAMAMOTO NTRENT JO ET AL.: "Synthesis of potent CXCR4 inhibitors possessing low cytotoxicity and improved biostability based on T140 derivatives", ORG BIOMOL CHEM, vol. 1, 2003, pages 3656 - 3662, XP008125664, DOI: 10.1039/B306473P
VITTORIO GEBBIA ET AL: "Irinotecan Plus Bolus/Infusional 5-Fluorouracil and Leucovorin in Patients With Pretreated Advanced Pancreatic Carcinoma : A Multicenter Experience of the Gruppo Oncologico Italia Meridionale", AMERICAN JOURNAL OF CLINICAL ONCOLOGY (CANCER CLINICAL TRIALS)., vol. 33, no. 5, 1 October 2010 (2010-10-01), US, pages 461 - 464, XP055461997, ISSN: 0277-3732, DOI: 10.1097/COC.0b013e3181b4e3b0 *

Similar Documents

Publication Publication Date Title
Kim et al. Liposomal encapsulation of a synergistic molar ratio of cytarabine and daunorubicin enhances selective toxicity for acute myeloid leukemia progenitors as compared to analogous normal hematopoietic cells
TWI791467B (en) Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluorouracil and leucovorin
AU2015249633B2 (en) Methods of treating early breast cancer with Trastuzumab-MCC-DM1 and Pertuzumab
CA2946538A1 (en) Use of dianhydrogalactitol and analogs or derivatives thereof to treat non-small-cell carcinoma of the lung and ovarian cancer
EP3821887A1 (en) Use of mitoxantrone liposome for treating non-hodgkin's lymphoma
WO2017172678A1 (en) Methods for treating cancer using combination therapies comprising an oligoclonal anti-egfr antibody preparation and lipsomal irinotecan
EP2638395A1 (en) Methods of treating cancer
US20230093147A1 (en) Methods and compositions relating to improved combination therapies
TW201922282A (en) Combination use of PD-1 antibody and epigenetic modulating agent in the preparation of a medicament for the treatment of tumor
Herzog et al. Trabectedin followed by irinotecan can stabilize disease in advanced translocation-positive sarcomas with acceptable toxicity
TW202231283A (en) Improved fluorouracil-based multi-agent chemotherapy for treatment of metastatic colorectal cancer
US9012422B2 (en) Method of treating acute myelogenous leukemia
US11406715B2 (en) Methods of treating HER2-positive metastatic breast cancer
WO2017176565A1 (en) Combinations of an anti-b7-h1 antibody and a cxcr4 peptide antagonist for treating a solid tumor
WO2020148744A1 (en) Combination therapy for treatment of pancreatic cancer
US20220117933A1 (en) Pharmaceutical Composition for Treating Tumor
EP3911353A1 (en) Specific combination therapy for treatment of pancreatic cancer
Löhr et al. Novel treatments and therapies in development for pancreatic cancer
ZA200508696B (en) Use of irinotecan for treatment of resistant breast cancer
US20220395497A1 (en) Cancer Treatment Using Camptothecin Derivatives
WO2021020336A1 (en) Pharmaceutical composition for treating tumor
Kitzen et al. Mild to moderate liver dysfunction does not require dose reduction of oral or intravenous vinorelbine: results of a pharmacokinetic study
Toldra American Association for Cancer Research (AACR)-110th Annual Meeting. Atlanta, Georgia, USA-March 29-April 3, 2019
CN116390735A (en) Combination of Bcl-2 inhibitors and hypomethylation agents for the treatment of cancer, uses and pharmaceutical compositions
KR20240029045A (en) Combination Treatment in Patients with Advanced and/or Metastatic Trop-2 Overexpressing Cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19795364

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19795364

Country of ref document: EP

Kind code of ref document: A1