WO2025219595A1 - Method for combination treatments using alkynylbenzenesulphonamides for cancer therapy - Google Patents
Method for combination treatments using alkynylbenzenesulphonamides for cancer therapyInfo
- Publication number
- WO2025219595A1 WO2025219595A1 PCT/EP2025/060790 EP2025060790W WO2025219595A1 WO 2025219595 A1 WO2025219595 A1 WO 2025219595A1 EP 2025060790 W EP2025060790 W EP 2025060790W WO 2025219595 A1 WO2025219595 A1 WO 2025219595A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- phenyl
- combination
- thiazol
- sulfonamido
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/425—Thiazoles
- A61K31/428—Thiazoles condensed with carbocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/337—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic 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
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/445—Non condensed piperidines, e.g. piperocaine
- A61K31/4523—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic 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
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4738—Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
- A61K31/4745—Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P33/00—Antiparasitic agents
Definitions
- the present disclosure pertains to novel cancer therapies.
- the present disclosure particularly relates to a combination of an anticancer treatment with a benzene sulfonamide thiazole compound for use in the treatment of cancer.
- Chemotherapy is often part of the first-line anticancer regimen. Although showing good results in a large number of cancers, it is also accompanied by several adverse effects that can strongly impair patients’ quality of life and resistance mechanisms that affect the efficacy of treatment. A close management of the doses and regimens applied is essential for optimizing the chances of remission and maintaining the patient’s well-being.
- Targeted therapies developed for specifically targeting genes and molecules directly involved in carcinogenesis and tumor growth have also proven extremely useful. These therapies are a form of personalized medicine and contrary to traditional chemotherapy, they do not “simply” target any rapidly dividing cell. Targeted therapies are therefore supposed to be better tolerated by patients. Unfortunately, these therapies only address a small subset of patients, and the identification of appropriate molecular targets is still in progress and the development of the corresponding drugs takes a lot of time.
- Immunotherapy is now a recognized and well-established therapeutic alternative for treating cancer. It groups together several different therapies, all based on stimulating the immune system of the patient in order to recognize and attack his or her disease. Immunotherapy using immune checkpoint modulators has revolutionized the oncology field (Robert, C. Nat Commun 1 1 , 3801 , 2020). Studies at the origin of the concept of this type of therapy led to James P. Allison and Tasuku Honjo winning the Nobel Prize in Medicine in 2018. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage resulting from immune responses.
- the immunogenicity of a tumor depends on its antigenicity and on several other immunomodulatory factors that are produced either by tumor cells or by host cells in the tumor microenvironment.
- a non- immunogenic tumor can thus not trigger an immune response, regardless the presence or absence of any inhibitory signal sent to immune cells.
- Non-immunogenic tumors are generally associated with poorer prognoses and hardly respond to most therapeutic strategies.
- the present inventors have shown that a very specific group of benzene sulfonamide thiazole compounds have the ability to remarkably potentiate the effects of anticancer treatments including chemotherapy, targeted therapies and immunotherapies. These very specific compounds particularly present a remarkable pharmacokinetic profile which allows providing a high efficacy with reduced toxicity. They allow triggering a strong immune cell death (ICD) response (including calreticulin cell surface exposure HMGB1 and AnnexinAI release, and extracellular ATP release in multiple cancer cell lines), which makes them perfect candidates for combination with other anticancer treatments such as immunotherapies.
- ICD immune cell death
- the present inventors have shown a synergy between the compounds according to the present invention and various anticancer treatments in multiple cancer cell lines.
- the present disclosure pertains to a combination of an anticancer treatment with a compound of formula (I):
- Ri represents H, methyl, phenyl or acetyl
- R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl
- the compound of formula (I) is the compound of formula (II):
- FIG. 1 Effects of BPR001 -615 (PB615): (A) CHOP-mediated cancer cell death induced by ER Stress Pathway, (B) CHOP-mediated Apoptosis and Autophagy, (C) Calreticulin cell surface exposure in mouse colorectal cell lines and human melanoma cell lines.
- Figure 6 Figure 6A: Effect of BPR001 -615 and trastuzumab on immune cell infiltrates.
- Figure 6B Effect of BPR001 -615 and trastuzumab on NK and Treg cells infiltrations
- Figure 7 Effect of BPR001 -615 and trastuzumab on NK and Treg cells infiltrations
- Figure 9 Comparable effect of combinations with two different compounds of formula (I) in cell lines.
- Figure A9. BPR001 -615 vs PB673 each in combination with Cisplatin at 48h in ovarian cell line (OVCAR.3).
- Figure 9B. BPR001 -615 vs PB673 each in combination with Irinotecan metabolite SN-38 at 48h in gastric cell line (Katolll).
- Figure 10 Superior effect of BPR001 -615 compared to HA15 in combination with different chemotherapies in cell lines.
- Figure 10A BPR001 -615 vs HA15 and Cisplatin at 48h in ovarian cell line (OVCAR.3).
- Figure 10B BPR001 -615 vs HA15 and Irinotecan metabolite SN-38 at 48h in gastric cell line (Katolll).
- Figure 10C BPR001 -615 vs HA15 and Docetaxel at 48h in gastric cell line (Katolll).
- ICD immunogenic cell death
- APCs antigen-presenting cells
- T cells T cells to induce immune-mediated tumor clearance.
- DAMPs danger-associated molecular patterns
- Immunogenic Cell Death is a unique response pattern of cell death that can provoke long- lasting antitumor immunity by inducing damage-associated molecular pattern (DAMP) signals such as surface exposure of calreticulin(CRT), release of ATP, and secretion of HMGB1 .1
- DAMP damage-associated molecular pattern
- the inventors Using various cancer cell lines models, and in vivo models, the inventors have shown that the specific benzene sulfonamide thiazole compounds of formula (I) have the ability to remarkably potentiate the effects of anticancer treatments including chemotherapy, targeted therapies and immunotherapies by triggering ICD markers to treatments that would trigger no or not enough ICD markers leading to DAMP signal. They have strong anticancer properties and target GRP78. Surprisingly, the effects obtained when combining these compounds with anticancer treatments is significantly higher than those obtained when using each treatment alone.
- the present inventors have further showed that such a combination therapy triggered remarkable cytotoxic effect, tumor growth inhibition and patient overall survival, in particular in GRP78 overexpressing tumors.
- the combination therapy provided a CHOP-mediated cytotoxic effect on cancer cells via endoplasmic-reticulum (ER) Stress induction that is significantly higher than that obtained with each treatment alone.
- ER endoplasmic-reticulum
- the present invention thus represents an extremely promising therapy for the treatment of cancers.
- the present disclosure pertains to a combination of an anticancer treatment with a compound of formula (I):
- Ri represents H, methyl, phenyl or acetyl
- R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl
- the term “combination”, “co-administration”, “combined administration” or “concomitant administration” preferably refers to a combined administration of at least two therapeutic agents, where a first agent, typically a compound of formula (I) is administered at the same time or separately within time intervals, with a second agent, in the same subject in need thereof, where these time intervals allow that the combined partners show a cooperative or synergistic effect for treating a disorder, e.g. a cancer. It is not intended to imply that the therapeutic agents must be administered at the same time and/or formulated for delivery together although these methods of delivery are within the scope described herein.
- the compound of formula (I) can be administered concurrently with or prior to, or subsequent to one or more other additional therapies or therapeutic agents.
- the terms are also meant to encompass treatment regimens in which the agents are not necessarily administered by the same route of administration.
- C2-C10 alkyl refers to a monovalent or divalent or trivalent, linear or branched, saturated hydrocarbon chain, comprising 2-10 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, tert-butyl-methyl, n-pentyl, n hexyl, n-heptyl, or n-octyl group.
- cycloalkyl refers to a saturated or unsaturated monocyclic or polycyclic system, such as a fused or bridged bicyclic system, comprising 3-12 carbon atoms, such as the cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantly, decalinyl, or norbornyl groups.
- an anticancer treatment/agent for use in the treatment of cancer wherein said anticancer agent is used in combination with a compound of formula (I) as defined above;
- kit-of-parts refers to a combined preparation wherein the active ingredients are physically separated for use in a combined therapy by simultaneous administration or sequential administration to the patient.
- the anticancer agent and the compound of formula (I) are administered to the patient in a separate form, either simultaneously, separately or sequentially in any order, for the treatment of cancer.
- kits of parts per se i.e. to a kit of parts comprising:
- the anticancer treatment is any anticancer agent selected from chemotherapies, targeted therapies, immunotherapies and combinations thereof.
- chemotherapeutics refer to chemical compounds that are effective in inhibiting tumor growth.
- chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaorarnide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including irinotecan and topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin
- calicheamicin especially calicheamicin (1 1 and calicheamicin 21 1 , see, e.g., Agnew Chem Inti. Ed. Engl. 33:183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholinodoxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino
- paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and docetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin, carboplatin oxaloplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-1 1 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and phannaceutically acceptable salts, acids or derivatives of any of the above
- antihormonal agents that act to regulate or inhibit honnone action on tumors
- anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY1 17018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
- the chemotherapeutic agent for use in the context of the present disclosure is selected from doxorubicin, toxoids such as paclitaxel and docetaxel, gemcitabine, anti-metabolites such as methotrexate and 5-fluorouracil (5-FU), platinum analogs such as cisplatin and oxaloplatin, and a camptothecin such as irinotecan and topotecan.
- Targeteted therapies refer to agents that act by blocking the growth of cancer cells by interfering with specific targeted molecules needed for carcinogenesis and tumor growth. Most targeted therapies are either small-molecule drugs or monoclonal antibodies. It is noteworthy that some targeted therapies can qualify as immunotherapeutic agents and/or chemotherapeutic agents.
- targeted therapies include Bortezomib, Braf inhibitors such as vemurafenib and dabrafenib, Cobimetinib, Imatinib, Gefitinib, Erlotinib Sorafenib, Sunitinib, Dasatinib, Lapatinib, Nilotinib, tamoxifen, janus kinase inhibitors such as Tofacitinib, ALK inhibitors such as Crizotinib, Bcl- 2 inhibitors such as Venetoclax, Obatoclax, navitoclax, and gossypol, PARP inhibitors such as olaparib, rucaparib, niraparib and talazoparib, PI3K inhibitors such as perifosine, Apatinib, Zoptarelin doxorubicin, MEK inhibitors such as trametinib, CDK inhibitors, Hsp90 inhibitors, H,
- the targeted therapy for use according to the present disclosure is selected from Bortezomib, Vemurafenib and Cobimetinib.
- Immunotherapy refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy, biological response modifier therapy and biotherapy.
- immunotherapeutic agents include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non-cytokine adjuvants.
- the immunotherapeutic treatment may consist of administering the patient with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells).
- immune cells T cells, NK, cells, dendritic cells, B cells
- allogenic and/or autologous CAR therapies such as CAR-T cells, CAR-NK cells and CAR-M cells.
- Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
- Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system.
- Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the nonspecific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines).
- Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines.
- Non-specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
- cytokines have found application in the treatment of cancer either as general non-specific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies.
- Suitable cytokines include, but are not limited to, interferons, interleukins and colonystimulating factors.
- Interferons include the common types of IFNs, IFN-alpha (IFN-a), IFN-beta (IFN-beta) and IFN-gamma (IFN-y).
- IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy.
- IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages.
- NK natural killer
- IFN-alpha Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation).
- Roferon Roche Pharmaceuticals
- Intron A Strecombinant IFN-alpha
- Interleukins include IL-2, IL-4, IL-1 1 and IL-12.
- Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL-12; Wyeth Pharmaceuticals).
- Interleukins, alone or in combination with other immunotherapeutics or with chemotherapeutics have shown efficacy in the treatment of various cancers including renal cancer (including metastatic renal cancer), melanoma (including metastatic melanoma), ovarian cancer (including recurrent ovarian cancer), cervical cancer (including metastatic cervical cancer), breast cancer, colorectal cancer, lung cancer, brain cancer, and prostate cancer.
- Colony-stimulating factors include granulocyte colony stimulating factor (G-CSF or filgrastim), granulocyte-macrophage colony stimulating factor (GM-CSF or sargramostim) and erythropoietin (epoetin alfa, darbepoietin).
- colony stimulating factors are available commercially, for example, Neupogen® (G-CSF; Amgen), Neulasta (pelfilgrastim; Amgen), Leukine (GM-CSF; Berlex), Procrit (erythropoietin; Ortho Biotech), Epogen (erythropoietin; Amgen), Arnesp (erytropoietin).
- Colony stimulating factors have shown efficacy in the treatment of cancer, including melanoma, colorectal cancer (including metastatic colorectal cancer), and lung cancer.
- Non-cytokine adjuvants suitable for use in the combinations of the present disclosure include, but are not limited to, Levamisole, alum hydroxide (alum), Calmette-Guerin bacillus (ACG), incomplete Freund's Adjuvant (IFA), QS-21 , DETOX, Keyhole limpet hemocyanin (KLH) and dinitrophenyl (DNP).
- Non-cytokine adjuvants in combination with other immuno- and/or chemotherapeutics have demonstrated efficacy against various cancers including, for example, colon cancer and colorectal cancer (Levimasole); melanoma (BCG and QS-21 ); renal cancer and bladder cancer (BCG).
- immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e. comprise immune system components that were generated external to the body.
- Cancer vaccines have been developed that comprise whole cancer cells, parts of cancer cells or one or more antigens derived from cancer cells. Cancer vaccines, alone or in combination with one or more immuno- or chemotherapeutic agents are being investigated in the treatment of several types of cancer including melanoma, renal cancer, ovarian cancer, breast cancer, colorectal cancer, and lung cancer.
- the immunotherapeutic treatment may consist of an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E. Dudley and Steven A. Rosenberg “Adoptive immunotherapy for cancer: harnessing the T cell response, Nature Reviews Immunology, Volume 12, April 2012.
- adoptive immunotherapy the patient’s circulating lymphocytes, or tumor infiltrated lymphocytes, are isolated in vitro, activated by lymphokines such as IL-2 or transuded with genes for tumor necrosis, and readministered (Rosenberg et al., 1988; 1989).
- the activated lymphocytes are most preferably the patient’s own cells that were earlier isolated from a blood or tumor sample and activated (or “expanded”) in vitro.
- Immunotherapeutic treatment as described herein includes allogenic and/or autologous CAR therapies which are well known in the art, particularly including, without limitations CAR-T cells, CAR-M cells and CAR-NK cells.
- Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor.
- Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject's immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
- Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present disclosure include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22.
- Monoclonal antibodies are used in the treatment of a wide range of cancers including breast cancer (including advanced metastatic breast cancer), colorectal cancer (including advanced and/or metastatic colorectal cancer), ovarian cancer, lung cancer, prostate cancer, cervical cancer, melanoma and brain tumors.
- breast cancer including advanced metastatic breast cancer
- colorectal cancer including advanced and/or metastatic colorectal cancer
- ovarian cancer lung cancer, prostate cancer, cervical cancer, melanoma and brain tumors.
- Other examples include immune check point inhibitors.
- immune checkpoint protein is widely known in the art and refers to a molecule that is expressed by T cells and that either turns up a signal (stimulatory checkpoint molecules) or turns down a signal (inhibitory checkpoint molecules).
- Immune checkpoints constitute immune checkpoint pathways such as the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al., 201 1 . Nature 480:480- 489).
- inhibitory checkpoint molecules include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO1 , KIR, PD-1 , LAG-3, TIM-3 TIGIT and VISTA.
- A2AR (the “Adenosine A2A receptor”) is considered as an important checkpoint in cancer therapy: the presence of adenosine in the immune microenvironment leads to an A2a-receptor activation, and induces a negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine.
- B7-H4 also called VTCN1 , is expressed by tumor cells and tumor-associated macrophages and is involved in tumor escape.
- BTLA (“B and T Lymphocyte Attenuator”), also referred to as CD272, has HVEM (Herpesvirus Entry Mediator) as its ligand.
- HVEM Herpesvirus Entry Mediator
- Surface expression of BTLA is gradually downregulated during differentiation of human CD8 + T cells from the naive to effector cell phenotype.
- Tumor specific human CD8 + T cells express high levels of BTLA.
- CTLA-4 Cytotoxic T-Lymphocyte-Associated protein 4
- CD152 also called CD152
- IDO1 Introleamine 2,3- dioxygenase 1
- IDO1 is a tryptophan catabolic enzyme - a immune inhibitory-related enzyme.
- IDO1 is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumor angiogenesis.
- KIR Killer-cell Immunoglobulin-like Receptor
- LAG3 Lymphocyte Activation Gene-3
- PD-1 Programmed Death 1
- This checkpoint is the target of pembrolizumab commercialized by Merck. Targeting PD-1 allows restoring immune function in the tumor microenvironment.
- TIM-3 (“T-cell Immunoglobulin domain and Mucin domain 3”), is expressed on activated human CD4 + T cells and regulates Th1 and Th17 cytokines. TIM-3 acts as a negative regulator of Th1/Tc1 function by triggering cell death upon interaction with its ligand, galectin-9.
- VISTA V-domain Ig suppressor of T cell activation
- TIGIT T cell immunoreceptor with Ig and ITIM domains
- NK Natural Killer Cells
- Immune checkpoint inhibitor or “checkpoint blockade cancer immunotherapy agent” has its general meaning in the art and refers to any compound inhibiting the function of an immune inhibitory checkpoint protein. Inhibition includes reduction of function and full blockade.
- Immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules. Preferred immune checkpoint inhibitors are antibodies that specifically recognize immune checkpoint proteins.
- the immune checkpoint inhibitor used in the context of the present disclosure is administered for enhancing the proliferation, migration, persistence and/or cytoxic activity of CD8 + T cells in the patient.
- CD8 + T cells are a subset of T cells which express CD8 on their surface. They are MHC class l-restricted, and function as cytotoxic T cells.
- CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in major histocompatibility complex class l-restricted interactions.
- the ability of the immune checkpoint inhibitor to enhance CD8 + T cell killing activity may be determined by any assay well known in the art. Typically said assay is an in vitro assay wherein CD8 + T cells are brought into contact with target cells (e.g. target cells that are recognized and/or lysed by CD8 + T cells).
- the immune checkpoint inhibitor of the present disclosure can be selected for its ability to increase specific lysis by CD8 + T cells by more than about 20%, preferably with at least about 30%, at least about 40%, at least about 50%, or more.
- Examples of protocols for classical cytotoxicity assays are conventional.
- the immune checkpoint inhibitor is an agent which blocks an immunosuppressive receptor expressed by activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1 , also known as PD-1 ), or by NK cells, like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1 ).
- CTL4 cytotoxic T lymphocyte-associated protein 4
- PDCD1 programmed cell death 1
- NK cells like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1 ).
- the checkpoint blockade cancer immunotherapy agent is an antibody.
- the checkpoint blockade cancer immunotherapy agent is an antibody selected from the group consisting of anti-PD1 antibodies, anti-PDL1 antibodies, anti-PDL2 antibodies, anti-CTLA4 antibodies, anti-TIM-3 antibodies, anti-LAG3 antibodies, anti-IDO1 antibodies, anti-TIG IT antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti-BTLA antibodies, and anti-B7H6 antibodies.
- anti PD-1 , anti PD-L1 and anti PD-L2 antibodies are described in US Patent Nos. 7,488,802; 7,943,743; 8,008,449; 8,168,757; 8,217,149, and PCT Published Patent Application Nos: WG03042402, WO2008156712, WO201008941 1 , WO2010036959, WO201 1066342,
- the PD-1 blockers include anti-PD-L1 antibodies (such as e.g. Atezolizumab, Avelumab or Durvalumab). In other embodiments, the PD-1 blockers include anti-PD-L2 antibodies.
- the PD-1 blockers include anti-PD-1 antibodies and similar binding proteins such as nivolumab (MDX 1106, BMS 936558, ONO 4538), a fully human lgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-LI and PD-L2; lambrolizumab (MK-3475 or SCH 900475), a humanized monoclonal lgG4 antibody against PD-1 ; CT-01 1 a humanized antibody that binds PD-1 ; AMP-224 is a fusion protein of B7-DC; an antibody Fc portion; BMS-936559 (MDX- 1 105-01 ) for PD-L1 (B7- H1 ) blockade.
- nivolumab MDX 1106, BMS 936558, ONO 4538
- a fully human lgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-LI and
- anti-CTLA-4 antibodies are described in US Patent Nos: 5,81 1 ,097; 5,81 1 ,097; 5,855,887; 6,051 ,227; 6,207,157; 6,682,736; 6,984,720; and 7,605,238.
- One anti-CDLA-4 antibody is tremelimumab, (ticilimumab, CP-675,206).
- the anti-CTLA-4 antibody is ipilimumab (also known as 10D1 , MDX-D010) a fully human monoclonal IgG antibody that binds to CTLA-4.
- lymphocyte activation gene-3 (LAG-3) inhibitors such as IMP321 , a soluble Ig fusion protein (Brignone et al., 2007, J. Immunol. 179:4202-421 1 ).
- B7 inhibitors such as B7-H3 and B7-H4 inhibitors.
- B7-H3 and B7-H4 inhibitors include B7 inhibitors, such as B7-H3 and B7-H4 inhibitors.
- the anti-B7-H3 antibody MGA271 Lio et al., 2012, Clin. Cancer Res. July 15 (18) 3834.
- TIM3 T-cell immunoglobulin domain and mucin domain 3
- the natural ligand of TIM-3 is galectin 9 (Gal9).
- TIM-3 inhibitor refers to a compound, substance or composition that can inhibit the function of TIM-3.
- the inhibitor can inhibit the expression or activity of TIM-3, modulate or block the TIM-3 signaling pathway and/or block the binding of TIM-3 to galectin-9.
- the immune checkpoint inhibitor is an Indoleamine 2,3-dioxygenase (IDO) inhibitor, preferably an IDO1 inhibitor. Examples of IDO inhibitors are described in WO 2014150677.
- IDO inhibitors include without limitation 1 -methyl-tryptophan (IMT), p- (3- benzofuranyl)-alanine, p-(3-benzo(b)thienyl)-alanine), 6-nitro-tryptophan, 6- fluoro-tryptophan, 4- methyl-tryptophan, 5 -methyl tryptophan, 6-methyl-tryptophan, 5-methoxy-tryptophan, 5 -hydroxy- tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-CI-indoxyl 1 ,3- diacetate, 9- vinylcarbazole, acemetacin, 5-bromo-tryptophan, 5-bromoindoxyl diacetate, 3- Amino- naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohydanto
- the IDO inhibitor is selected from 1 -methyl-tryptophan, p-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3-Amino-naphtoic acid and p-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof.
- the immune checkpoint inhibitor is an anti-TIG IT (T cell immunoglobin and ITIM domain) antibody.
- the anticancer treatment according to the present disclosure is an immune checkpoint inhibitor, preferably selected from an anti-PD-1 antibody and an anti-PD-L1 antibody.
- the checkpoint blockade cancer immunotherapy agent is a PD-1 blocking antibody, such as Nivolumab or Pembrolizumab.
- the compound of formula (I) is selected from the group consisting of:
- N-(4-(3-((4-(Hex-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB614”;
- N-(4-(3-((4-(3-Hydroxyprop-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB608”
- PB612 N-(4-(3-((4-(Cyclohexylethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide
- N-(4-(3-((4-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB610”;
- N-(4-(3-((4-((trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB671 ”;
- N-(4-(3-((4-(dec-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB672”;
- N-(4-(3-((4-(4-phenylbut-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB673”;
- N-(4-(3-((3-(Oct-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB617”;
- PB620 N-(4-(3-((3-((Trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB620”;
- N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide herein referred to as “PB621 ”;
- PB622 N-(3-(2-Aminothiazol-4-yl)phenyl)-4-(oct-1 -yn-1 -yl)benzenesulfonamide, herein referred to as “PB622”.
- compositions of formula (I) according to the present disclosure can be in the form of pharmaceutically acceptable salts.
- Pharmaceutically acceptable salts include the acid addition and base salts thereof.
- Suitable acid addition salts are formed from acids, which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate
- Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, 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.
- suitable salts see "Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley- VCH, Weinheim, Germany, 2002).
- solvate describes a molecular complex comprising the benzene sulfonamide thiazole compound and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
- solvent molecules for example, ethanol.
- hydrate is employed when said solvent is water.
- complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts.
- complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts.
- the resulting complexes may be ionised, partially ionized, or non-ionized.
- the compounds of formula (I) thus include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof.
- the compounds of formula (I) include all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) and isotopically- labeled compounds of formula (I).
- the anticancer treatment and the compound of formula (I) according to the present disclosure are used for the treatment of cancer in a patient.
- the present disclosure thus provides a method for treating cancer comprising administering, to a patient in need thereof, a therapeutically effective amount of an anticancer treatment and of the compound of formula (I), particularly a compound of formula (II), as defined above.
- the present disclosure further provides the compound of formula (I), particularly a compound of formula (II), for its use in the manufacture of a medicament for treating cancer, either as monotherapy or in the disclosed combinations.
- the anticancer treatment and the compound of formula (I), particularly a compound of formula (II), are administered to the patient either simultaneously, separately or sequentially in any order.
- the anticancer treatment is administered after the compound of formula (I), particularly a compound of formula (II).
- the anticancer treatment is administered to a patient who has already received the compound of formula (I), particularly a compound of formula (II).
- the compound of formula (I), particularly a compound of formula (II) is administered after the anticancer treatment.
- the compound of formula (I) is administered to a patient who has already received the anticancer treatment.
- the terms “Subject” and “Patient” refer to a human or an animal suffering from cancer.
- the patient is a mammal.
- the patient can e.g. be a human, a feline such as a cat, a canine such as a dog or an equid such as a horse.
- the patient is human.
- references herein to "treatment” include references to curative, palliative and prophylactic treatment.
- a “treatment” aims at reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or reversing, alleviating, inhibiting the progress of, or preventing one or more symptoms of the disorder or condition to which such term applies.
- the term treatment when referring to cancer disorders, may refer to slowing or preventing the growth of a tumor, or reducing the size of a tumor, or eradicating a tumor, or preventing metastatic development.
- cancer refers to the physiological condition in subjects that is characterized by unregulated or dysregulated cell growth or death.
- cancer includes solid tumors and blood born tumors.
- the combination for use according to the present disclosure applies to various organs of cancer origin (such as breast, colon, gastric, rectum, pancreatic, lung, skin, head and neck, bladder, ovary, prostate), and also to various cancer cell types (adenocarcinoma, squamous cell carcinoma, large cell cancer, melanoma, etc).
- organs of cancer origin such as breast, colon, gastric, rectum, pancreatic, lung, skin, head and neck, bladder, ovary, prostate
- cancer cell types adenocarcinoma, squamous cell carcinoma, large cell cancer, melanoma, etc.
- the patient suffers from a solid cancer selected from the group consisting of skin cancer (e.g. melanoma, nonmelanoma skin cancer), colorectal cancer, adrenal cortical cancer, anal cancer, bile duct cancer (e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer), bladder cancer, bone cancer (e.g. osteoblastoma, osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma), sarcomas such as liposarcoma and soft-tissue sarcoma, brain and central nervous system cancer (e.g.
- skin cancer e.g. melanoma, nonmelanoma skin cancer
- colorectal cancer e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer
- bladder cancer e.g. osteoblastoma, osteosarcoma
- breast cancer e.g. ductal carcinoma in situ, infiltrating ductal carcinoma, infiltrating lobular carcinoma, lobular carcinoma in situ
- cervical cancer e.g., endometrial cancer
- esthesioneuroblastoma midline granuloma
- nasopharyngeal cancer neuroblastoma
- oral cavity and oropharyngeal cancer ovarian cancer, pancreatic cancer, penile cancer, pituitary cancer, prostate cancer, retinoblastoma, rhabdomyosarcoma (e.g. embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, pleomorphic rhabdomyosarcoma), salivary gland cancer, stomach cancer, testicular cancer (e.g. seminoma, nonseminoma germ cell cancer), thymus cancer, thyroid cancer (e.g. follicular carcinoma, anaplastic carcinoma, poorly differentiated carcinoma, medullary thyroid carcinoma), vaginal cancer, vulvar cancer, and uterine cancer (e.g. uterine leiomyosarcoma).
- rhabdomyosarcoma e.g.
- the patient suffers from a hematological cancer such as leukaemia, lymphoma (such as Hodgkin lymphoma or non-Hodgkin lymphoma) and myeloma.
- a hematological cancer such as leukaemia, lymphoma (such as Hodgkin lymphoma or non-Hodgkin lymphoma) and myeloma.
- the compounds according to the present disclosure have been shown to have high potency against sensitive and resistant cancer cell lines from melanoma and colorectal cancer.
- the cancer treated according to the present disclosure is selected from skin cancer and colorectal cancer.
- the cancer treated according to the present disclosure is a colorectal cancer, a gastric cancer, a pancreatic cancer, a breast cancer, a lung cancer or skin cancer (preferably melanoma).
- the present inventors submit that the compounds of formula (I) described herein render the tumor immunogenic, i.e. detectable by the immune system, thereby allowing for the potentiation of the anticancer effect of the combined anticancer treatment.
- the cancer according to the present disclosure is a non-immunogenic tumor.
- non-immunogenic tumor it is herein referred to a tumor that does not elicit T-cell response.
- the skilled person is familiar with this notion and knows how to determine whether a tumor is immunogenic or not (see e.g. Wang et al. Elife 8 (2019): e49020).
- Such tumors are generally associated with:
- the cancer according to the present disclosure is “resistant” to immunotherapy meaning that the patient does not or poorly respond to immunotherapy, and particularly to an immune checkpoint inhibitor monotherapy.
- the term “responder” refers to a patient that will achieve a response, i.e. a patient where the cancer is eradicated, reduced or stabilized after treatment.
- a non-responder or refractory patient includes patients for whom the cancer does not show reduction or stabilization after the immunotherapy, and particularly after the immune checkpoint therapy.
- the present inventors have further showed that the combination therapy according to the present disclosure triggers remarkable tumor cell inhibition in cancer lines from tumors overexpressing GRP78.
- pancreatic cancer Tong et al. Pancreatology 21 .7 (2021 ): 1378-1385) or colorectal cancer (Thornton et al. International journal of cancer 133.6 (2013): 1408-1418).
- the combination according to the present disclosure allows significantly reducing GRP78 levels.
- the combination therapy according to the present disclosure can thus be particularly beneficial to patients presenting high levels of GRP78 and who are therefore identified as having a poor prognosis.
- the patient according to the present disclosure overexpresses GRP78.
- GRP78 also known as “Binding immunoglobulin protein” (BiP) or “heat shock 70 kDa protein 5” (HSPA5) is a protein that in humans is encoded by the HSPA5 gene. It is an endoplasmic reticulum chaperone that plays a key role in protein folding and quality control in the endoplasmic reticulum lumen.
- the sequence of GRP78 is available under reference P1 1021 in the Uniprot database library.
- control sample can be a sample obtained from a control population or from a control tissue.
- a control population is typically constituted of healthy subjects, i.e. subjects who do not suffer from cancer or any other disease.
- a control tissue is typically constituted of healthy tissues, i.e. tissues not affected by a disease. Said control tissue is usually obtained from the patient himself.
- the skilled person knows several techniques that allow determining whether a gene/protein is overexpressed as compared to a control sample. The skilled person is familiar with such techniques which are used routinely.
- the level of GRP78 is considered as “overexpressed” when said level is 1 .5, preferably 1 .7, more preferably 2 times higher than the level measured in a control population.
- the GRP78 level measured is either the circulating GRP78 level or the intratumor GRP78 level.
- “Circulating GRP78” refers to the GRP78 protein present in the blood circulation of the patient, typically in the cell-free section of the blood (plasma/serum) of the patient.
- the level of circulating GRP78 protein is typically measured by evaluating the quantity of GRP78 in a blood sample, typically a plasma/serum sample obtained from the patient.
- the control used for determining whether a level of circulating GRP78 is overexpressed is typically the GRP78 level measured in blood samples from a control population.
- “Intratumor GRP78” refers to the GRP78 protein expressed within the tumor of the patient.
- the level of the GRP78 protein is typically measured by evaluating the quantity of GRP78 expressed in a tumor sample, typically in a tumor biopsy obtained from the patient.
- the control used for determining whether the level of intratumor GRP78 is overexpressed is typically the GRP78 level measured in control tissues, i.e. healthy tissues from the patient.
- Such methods typically involve contacting the biological sample to be analyzed with an agent capable of specifically binding the target protein.
- This agent is usually a polyclonal or monoclonal antibody.
- the presence of the protein is then typically detected by standard immunodetection methods after separation of the proteins by electrophoresis (technique also called “Western blotting") or by immunoassays by direct, indirect, competition or immunocapture methods (techniques also called “ELISA”).
- the formation of a complex between the protein of interest and the antibody(s) targeting said protein is usually detected and quantified by measuring an enzymatic reaction generating a colored, chemiluminescent or fluorescent product leading in a specific staining pattern with a staining %.
- strong staining is defined as >50% of the tumor cells staining positive
- moderate staining is defined as 10 to ⁇ 50% of the tumor cells staining positive
- weak staining is defined as ⁇ 10% of the tumor cells staining positive (Samanta et al, 2020).
- kits are currently available for measuring the plasma/serum level GRP78 level.
- the ELISA kit commercialized by Enzo Life Sciences under reference ADI-900-214 can be used.
- the intratumor GRP78 level by determining the density of cells expressing GRP78.
- methods for measuring the density GRP78 expressing cells comprise a step of contacting the tumor tissue sample with at least one selective binding agent capable of selectively interacting with GRP78.
- the selective binding agent may be a polyclonal antibody or a monoclonal antibody, an antibody fragment, synthetic antibodies, or other protein-specific agents such as nucleic acid or peptide aptamers.
- the skilled person knows several antibodies which are specific to GRP78. Many of these antibodies are commercially available. Immunohistochemistry is particularly suitable for evaluating the density of GRP78 cells.
- the tissue tumor sample is firstly incubated with labelled antibodies directed against GRP78.
- the level of GRP78 can also be measured by determining the quantity of mRNA produced by the HSPA5 gene. Methods for determining a quantity of mRNA are well known in the art. For example, nucleic acid contained in the samples is first extracted according to standard methods, for example using lytic enzymes or chemical solutions or extracted by nucleic-acid-binding resins following the manufacturer's instructions. The extracted mRNA is then detected by hybridization (e. g., Northern blot analysis) and/or amplification (e.g., RT-PCR). Quantitative or semi-quantitative RT- PCR are preferred methods.
- hybridization e. g., Northern blot analysis
- amplification e.g., RT-PCR
- the present disclosure provides a method for treating cancer in a patient comprising a step of measuring the level of GRP78 in a tumor or blood/plasma/serum sample obtained from said patient, and then a step of administering a therapeutically effective amount of an anticancer treatment and of a compound of formula (I), particularly a compound of formula (II), as defined above if said patient is identified as overexpressing GRP78.
- the compounds used in the context of the present disclosure may be administered by any suitable route.
- the skilled person knows which route of administration to use as well as the corresponding dosages.
- Compounds useful in the context of the present disclosure are typically administered via parenteral (e.g., intravenous, intramuscular or subcutaneous) or via oral administration.
- Another aspect of the disclosure is a compound of Formula (I): Formula (I) wherein
- Ri represents H, methyl, phenyl or acetyl
- R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4, for use in the treatment of a GRP78 overexpressing tumor in a patient in need thereof.
- a “GRP78 overexpressing tumor” means a tumor from a patient characterized by a positive staining of GRP78 for at least 1 %, preferably at least 10% of the tumor cells in a tumor sample. Such staining may be measured by methods well known in the art and particularly as described in Samanta et al., 2020.
- a “GRP78 overexpressing tumor” may be characterized by measuring GRP78 expression at intratumor or circulating level as described above.
- the compounds of formula (I), particularly the compound of formula (II), either in monotherapy or in combination treatment as disclosed herein, is useful for patients having GRP78 overepressing tumor, whether high GRP78 tumor or low GRP78 tumor.
- the compound of formula I or formula II is for use in the treatment of a GRP78 positive tumor in patient in need thereof, wherein said tumor is a high GRP78 tumor.
- high GRP78 tumor it is meant a tumor from a patient characterized by a positive staining of GRP78 for at least 50% of the tumor cells present in a sample obtained from said tumor.
- the compound of formula I or formula II is for use in the treatment of a GRP78 positive tumor in patient in need thereof, wherein said tumor is a low GRP78 tumor.
- low GRP78 tumor a tumor from a patient characterized by a positive staining of GRP78 for 1 % to less than 50%, preferably 10% to less than 50%, of the tumor cells present in a sample obtained from said tumor.
- the compound of formula I is for use in the treatment of a GRP78 overexpressing tumor in patient in need thereof, wherein said patient has a low GRP78 tumor and wherein the compound of formula (I) is the compound BPR001 -615 of formula (II) described above.
- the patient to be treated with the compound of formula I, particularly compound BPR001 -615 of formula (II) is having a low GRP78 tumor, i.e. a patient tumor with GRP78 staining by immunohistochemistry shows a staining with 1 % to less than 50%, preferably 10% to less than 50%, of positive tumor cells, as determined according to Samanta et al. 2020, meaning that between 1 % to less than 50%, preferably 10% to less than 50%, of the tumor cells staining is positive, as described in Samanta et al, 2020.
- the patient to be treated with the combination is having a high GRP78 tumor, i.e. a patient tumor with GRP78 staining by immunohistochemistry shows a staining with at least 50% of positive tumor cells, as determined according to Samanta et al. 2020, meaning that between 50% to less than 100% of the tumor cells staining is positive, as described in Samanta et al, 2020.
- the compound of formula (I), particularly a compound of formula (II), as described herein is for use in the treatment of a GRP78 positive tumor, either a low GRP78 tumor or a high GRP78 tumor, in a patient in need thereof either alone as monotherapy or in combination with an anticancer agent, such combination being described in detail in the present disclosure.
- Methanol, ethanol, pyridine, DMF, ethyl acetate, diethyl ether and dichloromethane were purchased from Sigma Aldrich. DMF was dried by distillation under reduced pressure over MgSCk, pyridine and triethylamine were distilled over CaH2 under reduced pressure while methanol, ethanol, diethyl ether, ethyl acetate and dichloromethane were used as received. All chemicals were purchased from Aldrich, Merck or Alfa Aesar and used without further purification.
- Thin layer chromatography was performed on precoated Merck 60 GF254 silica gel plates and revealed first by visualization under UV light (254 nm and 365 nm) then spraying (ninhydrin or F SO EtOH).
- 1 H and 13 C NMR spectra were recorded on a Bruker Advance 200 MHz spectrometer or a Bruker Advance 400 MHz.
- Mass spectra (ESI-MS) were recorded on a Bruker (Daltonics Esquire 3000+).
- HRMS spectra were recorded on a ThermoFisher Q Exactive (ESI-MS) at a resolution of 140 000 at m/z 200.
- N-(4-(3-((4-(Oct-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (BPR001-615).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (400.0 mg, 0.88 mmol), Pd(PPhs)4 (152.5 mg, 15% mol.), copper (I) iodide (25.1 mg, 15%mol) and oct-1 -yne (653.2 pL, 4.43 mmol).
- n-Hex-1 -yne (188.0 pL, 1 .66 mmol) was added to the medium and the resulting mixture was stirred for 2h at 80°C (TLC monitoring). The reaction mixture was cooled to r.t. and all volatiles were removed under reduced pressure. Purification by silica gel column chromatography (CH2Cl2:EtOAc; 100:0 to 70:30) afforded alkyne 7 as an off-white powder (1 17.2 mg, 78%).
- N-(4-(3-((4-(3-Hydroxyprop-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB608).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (300.0 mg, 0.66 mmol), Pd(PPhs)4 (1 14.4 mg, 15% mol.), copper (I) iodide (18.9 mg, 15% mol.) and propargyl alcohol (190.4 pL, 3.30 mmol).
- N-(4-(3-((4-(Cyclohexylethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB612).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (1 10 mg, 0.24 mmol), Pd(PPhs)2Cl2 (25.7 mg, 15% mol.), copper (I) iodide (7.0 mg, 15% mol.) and cyclohexylacetylene (159.4 pL, 1 .22 mmol).
- N-(4-(3-((4-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB610).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (300.0 mg, 0.66 mmol), Pd(PPhs)4 (1 14.4 mg, 15% mol.), copper (I) iodide (18.9 mg, 15% mol.) and trimethylsilylacetylene (281 .9 pL, 1 .98 mmol).
- N-(4-(3-((4-((trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB671).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (22.0 mg, 7.5% mol.), copper (I) iodide (5.0 mg, 10% mol.) and trimethylsilylacetylene (178.0 pL, 1 .25 mmol).
- N-(4-(3-((4-(dec-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB672).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (339.0 mg, 0.75 mmol), Pd(PPhs)4 (66.0 mg, 7.5% mol.), copper (I) iodide (15.0 mg, 10% mol.) and dec-1 -yne (677.0 pL, 3.75 mmol).
- N-(4-(3-((4-(4-phenylbut-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB673).
- the general procedure for Sonogashira coupling was followed using bromoaryle 5a (339.0 mg, 0.75 mmol), Pd(PPhs)4 (66.0 mg, 7.5% mol.), copper (I) iodide (15.0 mg, 10% mol.) and dec-1 - yne (677.0 pL, 3.75 mmol).
- N-(4-(3-((3-(Oct- 1 -yn- 1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB617).
- the general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and oct-1 -yne (185.0 pL, 1 .25 mmol).
- N-(4-(3-((3-(3-Hydroxyprop-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB619).
- the general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and propargyl alcohol (72.0 pL, 1 .25 mmol).
- N-(4-(3-((3-((Trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB620).
- the general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and trimethylsilylacetylene (178.0 pL, 1 .25 mmol).
- N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB621).
- PB621 N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide
- N-(3-(2-(Methylamino)thiazol-4-yl)phenyl)-4-(oct- 1 -yn- 1 -yl)benzenesulfonamide (PB611 ).
- the Sonogashira coupling was performed using 5b (325.0 mg, 0.77 mmol), Pd(PPh 3 ) 2 CI 2 (80.6 mg, 15% mol.), copper (I) iodide (21 .9 mg, 15% mol.) and oct-1 -yne (565.0 pL, 3.83 mmol).
- N-(3-(2-Aminothiazol-4-yl)phenyl)-4-(oct-1-yn-1-yl)benzenesulfonamide (PB622).
- a suspension of 1 in 2M aq. HCI/EtOH (1 mL/1 mL) was stirred 2h at 80°C, then 15h at 50°C.
- the mixture was partitioned between EtOAc and saturated aq. Na 2 CO3, the aqueous phase was extracted with EtOAc once.
- the combined organic layers were dried with Na 2 SO4 and concentrated under reduced pressure to afford the pure amine 23 as a pale yellow solid (20.8 mg, 99%).
- Cell lines and reagents The different cell lines were purchased from ATCC. The tumor cell lines were maintained at 37C° and 5% CO2 in humidified atmosphere and grown in DMEM, high glucose, GlutaMAXTM Supplement, pyruvate growth media supplemented with 10% of fetal bovine serum, (ThermoFisher) under normal growing conditions. Cells were treated with the indicated anticancer agent and/or BPR001 -615 at the indicated concentrations and time. All drugs were dissolved in DMSO.
- Proliferation analysis Cell proliferation was measured using WST-1 reagent from Abeam (#ab65473). At Day 0, cells were plated in 96-well tissue culture plate. At day 1 , the cells were starved in serum (100pL/well). At Day 2 cells were treated with different drugs or DMSO at the indicated concentrations, in quadruplicates. 48h post treatment, WST-1 reagent (10pL/well) was added. The plate was read at TO at 450 nm on a Multiskan FC Counter (ThermoScientific), then every hour until O.D reach 1 .0. Cell proliferation is expressed as percent of the absorbance after subtraction to background (TO).
- Antibodies and Reagents Antibody against HMGB1 (#Ab18256), antibody against Annexin A1 (#Ab214486) and antibody against Calreticulin (#Ab2907) were purchased from Abeam (Paris, France).
- Anti-rabbit IgG and HRP-linked Antibody (#7074S) were purchased from Cell Signaling (Paris, France).
- Secondary antibody Texas Red goat (#T6391 ) was purchased from ThermoFisher (Paris, France).
- Oxaliplatin (OXP, Cas 61825-94) and Mitoxanthrone (MTX, Cas 70476-82) were used to induce immunogenic cell death.
- Fixable viability stain 510 (#564406) was purchased from BD Horizon (Paris, France).
- HMGB1 Release A375 and CT26 cells were plated at 180 000 cells/ml in 6 wellplates in medium with 2% FBS. The medium was changed after 24h and treatment was added to the cells. Oxaliplatin (OXP) was used at 50pM and 10OpM and BPR001 -615 was used at 3pM, 5pM and 7.5pM. 48h post treatment, supernatants were collected, dying tumor cells were removed by centrifugation and supernatants were isolated and frozen immediately at -20°C. Quantification of HMGB1 and Annexin A1 in the supernatants were assessed by western blot by loading 45pl of the supernatant supplementing with loading buffer. BSA detectable by ponceau staining was used as a loading control.
- Detection of ATP Release A375 cells and U2OS cells were plated at 10 000 cells/ml in 96 well plates with medium supplemented with 2% FBS. 24h later, treatment was added onto the cells (50pL/well) according to the manufacturer’s instructions. Mitoxanthrone (MTX) was used as positive control at 5pM, 10pM and 12.5pM. BPR001 -615 was used in dose response from 2.5pM to 15pM. Following the treatment, RealTime-Glo Extracellular ATP Assay Reagent was added. Mix plate by orbital shaking (300-500rpm) for 30-45 seconds to ensure homogeneity. Quantification of ATP release was assessed by RealTime-Glo Extracellular ATP Assay (Promega, #GA5010).
- BPR001 -615 alone has cytotoxic effects on cancer cell lines associated with ICD properties triggering the 3 hallmarks of ICD in dose-response:
- BPR001 -615 is a BiP(GRP78) Inhibitor inducing ER Stress and CHOP induction ( Figure 1 A).
- BPR001 -615 triggers ER Stress-induced & CHOP mediated cancer cells death ( Figure 1 B).
- BPR001 -615 triggers calreticulin cell surface exposure on multiple cancer cell lines ( Figure 1 C).
- BPR001 -615 triggers HMGB1 and AnnexinAI release ( Figure 2A).
- BPR001 -615 triggers extracellular ATP release on multiple cancer cell lines ( Figure 2B).
- Table 1 Summary of in vitro combination synergy observed at BPR001 -615 concentration with various chemotherapies in gastric adenocarcinoma cancers. All combinatory effects in chemotherapy dose response are presented in Figure 3.
- NXG mice A total of 40 female NXG mice aged 5-8 weeks were used for the study (Janvier Laboratories). Mice were allowed 7-days acclimatization before entering the study. Animals were housed in IVC cages (up to 5 per cage) with individual mice identified by tail mark. All animals were allowed free access to a standard certified commercial diet and sanitized water during the study. The holding room was maintained under standard conditions: 20-24° C, 45-65% humidity and a 12h light/dark cycle. NCI-N87 cells (5 x106 1 :1 with matrigel) were implanted on the rear dorsum of female NXG mice.
- mice/group 7 mice/group: (i) vehicle (15% Kolliphor HS15, 10% PEG400, 5% Ethanol, 70% ultrapure water); (ii) BPR001 -615 (150 mg/kg, Per Os twice a day (BiD) - 12hrs gap); (iii) Trastuzumab 10mg/kg, intravenous (IV), day 1 and day 16; (iv) combination BPR001 -615 150 mg/kg and Trastuzumab 10mg/kg.
- PBMCs from 2 donors were isolated, processed and dosed IV to the animals immediately prior to therapeutic dosing on the same day as treatment to maximize the treatment window time.
- Tumors were measured three times per week using digital calipers. The length, width and depth of the tumor were measured and used to calculate the tumor volume. The bodyweight of all mice on the study were also measured and recorded three times weekly.
- BPR001 -615 150mg/kg BID alone or in combination with trastuzumab was well tolerated.
- BPR001 -615 alone or in combination with trastuzumab decreased intratumoral GRP78 concentrations, with a more significant decrease observed when the drugs are used in combination (see Figure 4).
- BPR001-615 induces tumor cell death in combination with other treatments
- a combination treatment regimen consisting of BPR001 -615 together with an anti-programmed cell death protein 1 antibody (a-PD1 ) was investigated in C57BL/6J female mice subcutaneously implanted with MC38 cancer cells.
- IP intraperitoneal injection
- the efficacy of BPR001 -615 in combination with a-PD1 was mainly assessed through survival and tumor size evolution. Results of the in vivo assessment are presented in Figure 8.
- the overall survival graph shows an improvement in all treatment groups.
- BPR001 -615 shows a superior effect compared to HA15, demonstrating the superiority of compounds of formula I in combination with anticancer agents compared to other compounds ( Figure 10).
- BPR001 -615 The efficacy of BPR001 -615 on gastric tumour growth was evaluated using PBMC-humanized NXG female mice that were subcutaneously implanted with the GC cell line NCI-N87 (1 x 107 in Matrigel 1 :1 ). When tumour volume reached around 150 mm 3 ), PBMCs from two donors were isolated, processed and dosed IV to the animals prior to therapeutic dosing. BPR001 -615 was administered orally twice a day with 12h between administration until day 28 at 150 mg/kg. The study was also conducted on 7 mice treated with the standard-of-care targeted therapy Trastuzumab (a monoclonal antibody targeting HER2), administered on day 1 and day 16.
- Trastuzumab a monoclonal antibody targeting HER2
- BPR001 -615 is as efficient as targeted therapy standard-of-care Trastuzumab administered as single agent. This efficacy is demonstrated in BiP (or GRP78) low expressing tumours extending the potential of patients susceptible to respond to the treatment, therefore, to address more than 50% of BiP positive patients, which is far higher than HER2-positive patients population targeted by Trastuzumab (-20%).
- plasmatic GRP78 can also be used as a biomarker for patients’ selection based on expression of plasmatic GRP78 (low or high).
Landscapes
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Public Health (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Tropical Medicine & Parasitology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure pertains to a combination of specific benzene sulfonamide thiazole compounds with an anticancer treatment for use in the treatment of cancer in a patient in need thereof. Using several cancer cell models, the present inventors have shown that a combined therapy using specific benzene sulfonamide thiazole compounds and an anticancer treatment such as an 5 immunotherapeutic agent, a chemotherapeutic agent or targeted therapies allows specifically triggering immune cell death markers, thereby potentializing the effects of each compound alone.
Description
Description
Title: Method for combination treatments using alkynylbenzenesulphonamides for cancer therapy
Technical Field
[0001] The present disclosure pertains to novel cancer therapies. The present disclosure particularly relates to a combination of an anticancer treatment with a benzene sulfonamide thiazole compound for use in the treatment of cancer.
Background Art
[0002] Cancer encompasses a large family of diseases characterized by uncontrolled cell growth and division. Together, the over 200 known forms of cancer inflict a terrible social burden in terms of loss of life, diminished quality of life, healthcare costs and reduced productivity. Significant improvements in the diagnosis, screening and treatment of cancer have been made in the last decade. Nevertheless, cancer is still a leading cause of death worldwide: it accounted for nearly 10 million deaths in 2022 (Global Cancer Observatory: Cancer Today).
[0003] Many types of cancer treatments have been developed and are available such as chemotherapy, radiotherapy and immunotherapy.
[0004] Chemotherapy is often part of the first-line anticancer regimen. Although showing good results in a large number of cancers, it is also accompanied by several adverse effects that can strongly impair patients’ quality of life and resistance mechanisms that affect the efficacy of treatment. A close management of the doses and regimens applied is essential for optimizing the chances of remission and maintaining the patient’s well-being.
[0005] Targeted therapies developed for specifically targeting genes and molecules directly involved in carcinogenesis and tumor growth have also proven extremely useful. These therapies are a form of personalized medicine and contrary to traditional chemotherapy, they do not “simply” target any rapidly dividing cell. Targeted therapies are therefore supposed to be better tolerated by patients. Unfortunately, these therapies only address a small subset of patients, and the identification of appropriate molecular targets is still in progress and the development of the corresponding drugs takes a lot of time.
[0006] Immunotherapy is now a recognized and well-established therapeutic alternative for treating cancer. It groups together several different therapies, all based on stimulating the immune system of the patient in order to recognize and attack his or her disease. Immunotherapy using immune checkpoint modulators has revolutionized the oncology field (Robert, C. Nat Commun 1 1 , 3801 , 2020). Studies at the origin of the concept of this type of therapy led to James P. Allison and Tasuku Honjo winning the Nobel Prize in Medicine in 2018. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage resulting from immune responses. When the checkpoint
and its ligand bind together, they send an “off” signal to the T cells, thereby inhibiting/strongly reducing immune responses. It is now clear that tumors use certain immune checkpoint pathways in order to escape immunity responses. Immune checkpoint inhibitors (ICI) work by blocking the checkpoint from binding with its ligand so as to prevent the “off” signal from being sent. These compounds have shown remarkable clinical efficacy and represent substantial hope for cure for some treatment-resistant tumors. Unfortunately, not all tumors respond to immune checkpoint inhibitors. In some cases, even after having turned off the inhibitory signal sent by immune checkpoints, the immune system is simply not capable of detecting and attacking the tumor. Some tumors are indeed non-immunogenic which means that they cannot be detected by immune cells. The immunogenicity of a tumor depends on its antigenicity and on several other immunomodulatory factors that are produced either by tumor cells or by host cells in the tumor microenvironment. A non- immunogenic tumor can thus not trigger an immune response, regardless the presence or absence of any inhibitory signal sent to immune cells. Non-immunogenic tumors are generally associated with poorer prognoses and hardly respond to most therapeutic strategies.
[0007] There is thus a continued and urgent need for novel anticancer therapies and for the identification of new molecules that would allow potentiating the effects of known anticancer treatments.
Summary
[0008] The invention is defined by the claims.
[0009] The present inventors have shown that a very specific group of benzene sulfonamide thiazole compounds have the ability to remarkably potentiate the effects of anticancer treatments including chemotherapy, targeted therapies and immunotherapies. These very specific compounds particularly present a remarkable pharmacokinetic profile which allows providing a high efficacy with reduced toxicity. They allow triggering a strong immune cell death (ICD) response (including calreticulin cell surface exposure HMGB1 and AnnexinAI release, and extracellular ATP release in multiple cancer cell lines), which makes them perfect candidates for combination with other anticancer treatments such as immunotherapies. The present inventors have shown a synergy between the compounds according to the present invention and various anticancer treatments in multiple cancer cell lines.
[0010] Accordingly, the present disclosure pertains to a combination of an anticancer treatment with a compound of formula (I):
[0011] [Formula I] :
[0012] wherein
[0013] Ri represents H, methyl, phenyl or acetyl;
[0014] R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and
[0015] wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4,
[0016] for use in the treatment of cancer in a patient in need thereof.
[0017] Particularly, the compound of formula (I) is the compound of formula (II):
[0018] [Formula II]:
Brief Description of Drawings
Figure 1
[0019] [Figure 1] Effects of BPR001 -615 (PB615): (A) CHOP-mediated cancer cell death induced by ER Stress Pathway, (B) CHOP-mediated Apoptosis and Autophagy, (C) Calreticulin cell surface exposure in mouse colorectal cell lines and human melanoma cell lines.
Figure 2
[0020] [Figure 2] Effects of BPR001 -615: (A) HMGB1 and Annexin A1 release, (B) Extracellular ATP release in human melanoma cell lines and osteosarcoma cell lines.
Figure 3
[0021] [Figure 3] BPR001 -615 synergizes with chemotherapies to induce cancer cell death of gastric cancer cell lines (Viability of cancer cells vs DMSO). WST-1 cell proliferation assay, Cell viability upon combination treatment of BPR001 -615 with chemotherapy. 2Way-ANOVA test, * p<0.05, ** p<0.01 , *** p< 0.005, SN-38: Irinotecan metabolites SN38, Oxa: Oxaliplatine.
Figure 4
[0022] [Figure 4] Effect of BPR001 -615 in combination with trastuzumab on NCI-N87 tumor model grown in PBMC-humanized NXG mice.
Figure 5
[0023] [Figure 5] Effect of BPR001 -615 and trastuzumab on GRP78 concentration on NCI-N87 tumor
Figure 6
[0024] [Figure 6] Figure 6A: Effect of BPR001 -615 and trastuzumab on immune cell infiltrates. Figure 6B: Effect of BPR001 -615 and trastuzumab on NK and Treg cells infiltrations
Figure 7
[0025] [Figure 7] The synergetic effect of BPR001 -615 with SN38 on cell death induction of CT26 cancer cell lines. WST-1 cell proliferation assay, Cell viability upon combination treatment of BPR001 -615 with chemotherapy. 2-Way ANOVA test, * p<0.05, ** p<0.01 , *** p<0.005, **** p<0.0001
Figure 8
[0026] [Figure 8] Survival curve after 49 days of treatment with BPR001 -615 alone and in combination with a-PD1 . mpk: mg/kg; *: Refer to significant survival difference between groups. Mice are sacrificed when tumour volume reaches a pre-determined volume.
Figure 9
[0027] [Figure 9] Comparable effect of combinations with two different compounds of formula (I) in cell lines. Figure A9. BPR001 -615 vs PB673 each in combination with Cisplatin at 48h in ovarian cell line (OVCAR.3). Figure 9B. BPR001 -615 vs PB673 each in combination with Irinotecan metabolite SN-38 at 48h in gastric cell line (Katolll).
Figure 10
[0028] [Figure 10] Superior effect of BPR001 -615 compared to HA15 in combination with different chemotherapies in cell lines. Figure 10A: BPR001 -615 vs HA15 and Cisplatin at 48h in ovarian cell line (OVCAR.3). Figure 10B: BPR001 -615 vs HA15 and Irinotecan metabolite SN-38 at 48h in gastric cell line (Katolll). Figure 10C. BPR001 -615 vs HA15 and Docetaxel at 48h in gastric cell line (Katolll).
Figure 11
[0029] [Figure 1 1 ] NCI-N87 tumour size evolution in PBMC-humanised NXG female mice treated with BPR001 -615 and Trastuzumab as single agents. Left panel: The statistical test used is 2-way ANOVA Test. Right panel: %tumour growth inhibition compared to Day 1 of treatment vs vehicle. GRP78 (BiP) overexpression in various cancer cell lines, NCI-N87 is highlighted (Western Blot analysis).]
Detailed description
[0030] Preclinical and clinical research in the past two decades has redefined the mechanism of action of some chemotherapeutics that are able to activate the immune system against cancer when cell death is perceived by the immune cells. This immunogenic cell death (ICD) activates antigen- presenting cells (APCs) and T cells to induce immune-mediated tumor clearance. One of the key requirements to achieve this effect is the externalization of the danger-associated molecular patterns (DAMPs), molecules released or exposed by cancer cells during ICD that increase the visibility of the cancer cells by the immune system (Cifric et al. Antioxidants and Redox Signaling (2024)).
[0031] Immunogenic Cell Death is a unique response pattern of cell death that can provoke long- lasting antitumor immunity by inducing damage-associated molecular pattern (DAMP) signals such as surface exposure of calreticulin(CRT), release of ATP, and secretion of HMGB1 .1 For cancer cells, the released immunostimulatory signals during the ICD process lead to the enhanced engulfment of
cancer cells by dendritic cells(DCs), the most efficient antigen-presenting cells in the immune system. Then, the cancer-specific antigens will be presented by DCs to the relevant T Cells, resulting in the activation of a global anticancer immune response.
[0032] Using various cancer cell lines models, and in vivo models, the inventors have shown that the specific benzene sulfonamide thiazole compounds of formula (I) have the ability to remarkably potentiate the effects of anticancer treatments including chemotherapy, targeted therapies and immunotherapies by triggering ICD markers to treatments that would trigger no or not enough ICD markers leading to DAMP signal. They have strong anticancer properties and target GRP78. Surprisingly, the effects obtained when combining these compounds with anticancer treatments is significantly higher than those obtained when using each treatment alone.
[0033] The present inventors have further showed that such a combination therapy triggered remarkable cytotoxic effect, tumor growth inhibition and patient overall survival, in particular in GRP78 overexpressing tumors.
[0034] The combination therapy provided a CHOP-mediated cytotoxic effect on cancer cells via endoplasmic-reticulum (ER) Stress induction that is significantly higher than that obtained with each treatment alone.
[0035] The specific compounds of formula (I) have cytotoxic effects on cancer cell lines associated with ICD properties triggering the 3 hallmarks of ICD in dose-response:
[0036] - Calreticulin exposure ;
[0037] - ATP release ; and
[0038] - HMGB1 release.
[0039] These compounds have two complementary and concomitant effects mediated by GRP78 inhibition and strong ER Stress induction that triggers 3 synergistic anti-tumoral effects: CHOP- mediated apoptosis and autophagy associated with ICD-mediated cell death and tumor destruction that potentiate chemotherapy, targeted therapy, radiotherapy and immunotherapy treatments.
[0040] The present invention thus represents an extremely promising therapy for the treatment of cancers.
[0041] Furthermore, the inventors have shown that the compounds of formula (I) have at least equivalent effects than well-known standard of care even in low GRP78 tumors (as defined below).
[0042] Accordingly, in a first aspect, the present disclosure pertains to a combination of an anticancer treatment with a compound of formula (I):
[0043] [Formula I] :
[0044] wherein
[0045] Ri represents H, methyl, phenyl or acetyl;
[0046] R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and
[0047] wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4,
[0048] for use in the treatment of cancer in a patient in need thereof.
[0049] As used herein, the term “combination”, “co-administration”, “combined administration” or “concomitant administration” preferably refers to a combined administration of at least two therapeutic agents, where a first agent, typically a compound of formula (I) is administered at the same time or separately within time intervals, with a second agent, in the same subject in need thereof, where these time intervals allow that the combined partners show a cooperative or synergistic effect for treating a disorder, e.g. a cancer. It is not intended to imply that the therapeutic agents must be administered at the same time and/or formulated for delivery together although these methods of delivery are within the scope described herein. The compound of formula (I) can be administered concurrently with or prior to, or subsequent to one or more other additional therapies or therapeutic agents. The terms are also meant to encompass treatment regimens in which the agents are not necessarily administered by the same route of administration.
[0050] The term “C2-C10 alkyl” refers to a monovalent or divalent or trivalent, linear or branched, saturated hydrocarbon chain, comprising 2-10 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, tert-butyl, tert-butyl-methyl, n-pentyl, n hexyl, n-heptyl, or n-octyl group.
[0051] The term “cycloalkyl” refers to a saturated or unsaturated monocyclic or polycyclic system, such as a fused or bridged bicyclic system, comprising 3-12 carbon atoms, such as the cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, adamantly, decalinyl, or norbornyl groups.
[0052] The “combination” for use according to the present disclosure encompasses both:
[0053] - an anticancer treatment/agent for use in the treatment of cancer, wherein said anticancer agent is used in combination with a compound of formula (I) as defined above; and
[0054] - a compound of formula (I) as defined above, e.g. BPR001 -615, for use in the treatment of cancer, wherein said compound is used in combination with an anticancer agent.
[0055] The “combination” for use according to the present disclosure also encompasses a “kit of parts” comprising:
[0056] - an anticancer agent; and
[0057] - a compound of formula (I) as defined above,
[0058] for use in the treatment of cancer.
[0059] The term “kit-of-parts” herein refers to a combined preparation wherein the active ingredients are physically separated for use in a combined therapy by simultaneous administration or sequential administration to the patient. Hence, according to the present disclosure, the anticancer agent and the compound of formula (I) are administered to the patient in a separate form, either simultaneously, separately or sequentially in any order, for the treatment of cancer.
[0060] According to a further embodiment, the present disclosure also pertains to a kit of parts per se, i.e. to a kit of parts comprising:
[0061] - an anticancer agent; and
[0062] - a compound of formula (I) as defined above, e.g., BPR001 -615.
[0063] Anticancer treatment/ Agent
[0064] According to the present disclosure, the anticancer treatment is any anticancer agent selected from chemotherapies, targeted therapies, immunotherapies and combinations thereof.
[0065] ‘Chemotherapies ”, “chemotherapeutics” and "chemotherapeutic agents" refer to chemical compounds that are effective in inhibiting tumor growth. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaorarnide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including irinotecan and topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estrarnustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimus tine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, especially calicheamicin (1 1 and calicheamicin 21 1 , see, e.g., Agnew Chem Inti. Ed. Engl. 33:183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholinodoxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idanrbicin, marcellomycin, mitomycins, mycophenolic acid, nogalarnycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptomgrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and
5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophospharnide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pento statin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; rhizoxin; sizofiran; spirogennanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylarnine; trichothecenes (especially T-2 toxin, verracurin A, roridinA and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobromtol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and docetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin, carboplatin oxaloplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-1 1 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and phannaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are antihormonal agents that act to regulate or inhibit honnone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY1 17018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
[0066] According to a preferred embodiment, the chemotherapeutic agent for use in the context of the present disclosure is selected from doxorubicin, toxoids such as paclitaxel and docetaxel, gemcitabine, anti-metabolites such as methotrexate and 5-fluorouracil (5-FU), platinum analogs such as cisplatin and oxaloplatin, and a camptothecin such as irinotecan and topotecan.
[0067] “Targeted therapies” refer to agents that act by blocking the growth of cancer cells by interfering with specific targeted molecules needed for carcinogenesis and tumor growth. Most targeted therapies are either small-molecule drugs or monoclonal antibodies. It is noteworthy that some targeted therapies can qualify as immunotherapeutic agents and/or chemotherapeutic agents. Examples of targeted therapies include Bortezomib, Braf inhibitors such as vemurafenib and dabrafenib, Cobimetinib, Imatinib, Gefitinib, Erlotinib Sorafenib, Sunitinib, Dasatinib, Lapatinib, Nilotinib, tamoxifen, janus kinase inhibitors such as Tofacitinib, ALK inhibitors such as Crizotinib, Bcl- 2 inhibitors such as Venetoclax, Obatoclax, navitoclax, and gossypol, PARP inhibitors such as olaparib, rucaparib, niraparib and talazoparib, PI3K inhibitors such as perifosine, Apatinib, Zoptarelin doxorubicin, MEK inhibitors such as trametinib, CDK inhibitors, Hsp90 inhibitors, Hedgehog pathway
inhibitors such as vismodegib and sonidegib, salinomycin VAL-083, Vintafolide, Temsirolimus, Everolimus, Vemurafenib, Trametinib, Dabrafenib, monoclonal antibodies including Pembrolizumab, Rituximab, Alemtuzumab, Cetuximab, Panitumumab, Bevacizumab and Ipilimumab.
[0068] According to a specific embodiment, the targeted therapy for use according to the present disclosure is selected from Bortezomib, Vemurafenib and Cobimetinib.
[0069] “Immunotherapy” “Immunotherapeutics” or "immunotherapeutic agent" refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy, biological response modifier therapy and biotherapy. Examples of common immunotherapeutic agents known in the art include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non-cytokine adjuvants. Alternatively, the immunotherapeutic treatment may consist of administering the patient with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells...), notably including, without limitation, allogenic and/or autologous CAR therapies such as CAR-T cells, CAR-NK cells and CAR-M cells.
[0070] Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
[0071] Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system. Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the nonspecific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines). Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines. Non-specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
[0072] A number of cytokines have found application in the treatment of cancer either as general non-specific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies. Suitable cytokines include, but are not limited to, interferons, interleukins and colonystimulating factors.
[0073] Interferons (IFNs) include the common types of IFNs, IFN-alpha (IFN-a), IFN-beta (IFN-beta) and IFN-gamma (IFN-y). IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy. IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting
the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages. Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation). The use of IFN-alpha, alone or in combination with other immunotherapeutics or with chemotherapeutics, has shown efficacy in the treatment of various cancers including melanoma (including metastatic melanoma), renal cancer (including metastatic renal cancer), breast cancer, prostate cancer, and cervical cancer (including metastatic cervical cancer).
[0074] Interleukins include IL-2, IL-4, IL-1 1 and IL-12. Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL-12; Wyeth Pharmaceuticals). Interleukins, alone or in combination with other immunotherapeutics or with chemotherapeutics, have shown efficacy in the treatment of various cancers including renal cancer (including metastatic renal cancer), melanoma (including metastatic melanoma), ovarian cancer (including recurrent ovarian cancer), cervical cancer (including metastatic cervical cancer), breast cancer, colorectal cancer, lung cancer, brain cancer, and prostate cancer.
[0075] Colony-stimulating factors (CSFs) include granulocyte colony stimulating factor (G-CSF or filgrastim), granulocyte-macrophage colony stimulating factor (GM-CSF or sargramostim) and erythropoietin (epoetin alfa, darbepoietin). Various-recombinant colony stimulating factors are available commercially, for example, Neupogen® (G-CSF; Amgen), Neulasta (pelfilgrastim; Amgen), Leukine (GM-CSF; Berlex), Procrit (erythropoietin; Ortho Biotech), Epogen (erythropoietin; Amgen), Arnesp (erytropoietin). Colony stimulating factors have shown efficacy in the treatment of cancer, including melanoma, colorectal cancer (including metastatic colorectal cancer), and lung cancer.
[0076] Non-cytokine adjuvants suitable for use in the combinations of the present disclosure include, but are not limited to, Levamisole, alum hydroxide (alum), Calmette-Guerin bacillus (ACG), incomplete Freund's Adjuvant (IFA), QS-21 , DETOX, Keyhole limpet hemocyanin (KLH) and dinitrophenyl (DNP). Non-cytokine adjuvants in combination with other immuno- and/or chemotherapeutics have demonstrated efficacy against various cancers including, for example, colon cancer and colorectal cancer (Levimasole); melanoma (BCG and QS-21 ); renal cancer and bladder cancer (BCG).
[0077] In addition to having specific or non-specific targets, immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e. comprise immune system components that were generated external to the body.
[0078] Active specific immunotherapy typically involves the use of cancer vaccines. Cancer vaccines have been developed that comprise whole cancer cells, parts of cancer cells or one or more antigens derived from cancer cells. Cancer vaccines, alone or in combination with one or more immuno- or chemotherapeutic agents are being investigated in the treatment of several types of cancer including melanoma, renal cancer, ovarian cancer, breast cancer, colorectal cancer, and lung cancer.
[0079] The immunotherapeutic treatment may consist of an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E. Dudley and Steven A. Rosenberg “Adoptive immunotherapy for
cancer: harnessing the T cell response, Nature Reviews Immunology, Volume 12, April 2012. In adoptive immunotherapy, the patient’s circulating lymphocytes, or tumor infiltrated lymphocytes, are isolated in vitro, activated by lymphokines such as IL-2 or transuded with genes for tumor necrosis, and readministered (Rosenberg et al., 1988; 1989). The activated lymphocytes are most preferably the patient’s own cells that were earlier isolated from a blood or tumor sample and activated (or “expanded”) in vitro. This form of immunotherapy has produced several cases of regression of melanoma and renal carcinoma. Immunotherapeutic treatment as described herein includes allogenic and/or autologous CAR therapies which are well known in the art, particularly including, without limitations CAR-T cells, CAR-M cells and CAR-NK cells.
[0080] Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor. Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject's immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
[0081] Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present disclosure include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22. Monoclonal antibodies are used in the treatment of a wide range of cancers including breast cancer (including advanced metastatic breast cancer), colorectal cancer (including advanced and/or metastatic colorectal cancer), ovarian cancer, lung cancer, prostate cancer, cervical cancer, melanoma and brain tumors. Other examples include immune check point inhibitors.
[0082] The expression "immune checkpoint protein" is widely known in the art and refers to a molecule that is expressed by T cells and that either turns up a signal (stimulatory checkpoint molecules) or turns down a signal (inhibitory checkpoint molecules). Immune checkpoints constitute immune checkpoint pathways such as the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al., 201 1 . Nature 480:480- 489). Examples of inhibitory checkpoint molecules include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO1 , KIR, PD-1 , LAG-3, TIM-3 TIGIT and VISTA.
[0083] A2AR (the “Adenosine A2A receptor”) is considered as an important checkpoint in cancer therapy: the presence of adenosine in the immune microenvironment leads to an A2a-receptor activation, and induces a negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine. B7-H3, also called CD276, was originally understood to be a co-stimulatory molecule but is now regarded as co-inhibitory. B7-H4, also called VTCN1 , is expressed by tumor cells and tumor-associated macrophages and is involved in tumor escape. BTLA
(“B and T Lymphocyte Attenuator”), also referred to as CD272, has HVEM (Herpesvirus Entry Mediator) as its ligand. Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype. Tumor specific human CD8+ T cells express high levels of BTLA. Expression of CTLA-4 (“Cytotoxic T-Lymphocyte-Associated protein 4”), also called CD152, on Treg cells controls T cell proliferation. IDO1 (“Indoleamine 2,3- dioxygenase 1 ”) is a tryptophan catabolic enzyme - a immune inhibitory-related enzyme. IDO1 is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumor angiogenesis. KIR (“Killer-cell Immunoglobulin-like Receptor”), is a receptor for MHC Class I molecules on Natural Killer cells. LAG3 (“Lymphocyte Activation Gene-3”) suppresses an immune response via an action on Tregs and a direct inhibitory effect on CD8+ T cells. PD-1 (“Programmed Death 1 ”) receptor, has two ligands, PD-L1 and PD-L2. This checkpoint is the target of pembrolizumab commercialized by Merck. Targeting PD-1 allows restoring immune function in the tumor microenvironment. TIM-3, (“T-cell Immunoglobulin domain and Mucin domain 3”), is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. TIM-3 acts as a negative regulator of Th1/Tc1 function by triggering cell death upon interaction with its ligand, galectin-9. VISTA (“V-domain Ig suppressor of T cell activation”) is primarily expressed on hematopoietic cells. Consistent expression of VISTA on leukocytes within tumors allow VISTA blockade to be effective across a broad range of solid tumors. TIGIT (“T cell immunoreceptor with Ig and ITIM domains”) is an immune receptor present on some percentage of T cells and Natural Killer Cells(NK). TIGIT inhibits T cell activation in vivo.
[0084] An “immune checkpoint inhibitor" or “checkpoint blockade cancer immunotherapy agent” has its general meaning in the art and refers to any compound inhibiting the function of an immune inhibitory checkpoint protein. Inhibition includes reduction of function and full blockade. Immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules. Preferred immune checkpoint inhibitors are antibodies that specifically recognize immune checkpoint proteins. The immune checkpoint inhibitor used in the context of the present disclosure is administered for enhancing the proliferation, migration, persistence and/or cytoxic activity of CD8+ T cells in the patient. CD8+ T cells are a subset of T cells which express CD8 on their surface. They are MHC class l-restricted, and function as cytotoxic T cells. They are also referred to as cytotoxic T lymphocytes (CTL), T-killer cell, cytolytic T cells, CD8+ T cells or killer T cells. CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in major histocompatibility complex class l-restricted interactions. The ability of the immune checkpoint inhibitor to enhance CD8+ T cell killing activity may be determined by any assay well known in the art. Typically said assay is an in vitro assay wherein CD8+ T cells are brought into contact with target cells (e.g. target cells that are recognized and/or lysed by CD8+ T cells). For example, the immune checkpoint inhibitor of the present disclosure can be selected for its ability to increase specific lysis by CD8+ T cells by more than about 20%, preferably with at least about 30%, at least about 40%, at least about 50%, or more. Examples of protocols for classical cytotoxicity assays are conventional.
[0085] Typically, the immune checkpoint inhibitor is an agent which blocks an immunosuppressive receptor expressed by activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein
4 (CTLA4) and programmed cell death 1 (PDCD1 , also known as PD-1 ), or by NK cells, like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1 ).
[0086] Typically, the checkpoint blockade cancer immunotherapy agent is an antibody.
[0087] In some embodiments, the checkpoint blockade cancer immunotherapy agent is an antibody selected from the group consisting of anti-PD1 antibodies, anti-PDL1 antibodies, anti-PDL2 antibodies, anti-CTLA4 antibodies, anti-TIM-3 antibodies, anti-LAG3 antibodies, anti-IDO1 antibodies, anti-TIG IT antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti-BTLA antibodies, and anti-B7H6 antibodies.
[0088] Examples of anti PD-1 , anti PD-L1 and anti PD-L2 antibodies are described in US Patent Nos. 7,488,802; 7,943,743; 8,008,449; 8,168,757; 8,217,149, and PCT Published Patent Application Nos: WG03042402, WO2008156712, WO201008941 1 , WO2010036959, WO201 1066342,
WO201 1 159877, WO201 1082400, and WO201 1 161699. In some embodiments, the PD-1 blockers include anti-PD-L1 antibodies (such as e.g. Atezolizumab, Avelumab or Durvalumab). In other embodiments, the PD-1 blockers include anti-PD-L2 antibodies. In certain other embodiments the PD-1 blockers include anti-PD-1 antibodies and similar binding proteins such as nivolumab (MDX 1106, BMS 936558, ONO 4538), a fully human lgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-LI and PD-L2; lambrolizumab (MK-3475 or SCH 900475), a humanized monoclonal lgG4 antibody against PD-1 ; CT-01 1 a humanized antibody that binds PD-1 ; AMP-224 is a fusion protein of B7-DC; an antibody Fc portion; BMS-936559 (MDX- 1 105-01 ) for PD-L1 (B7- H1 ) blockade.
[0089] Examples of anti-CTLA-4 antibodies are described in US Patent Nos: 5,81 1 ,097; 5,81 1 ,097; 5,855,887; 6,051 ,227; 6,207,157; 6,682,736; 6,984,720; and 7,605,238. One anti-CDLA-4 antibody is tremelimumab, (ticilimumab, CP-675,206). In some embodiments, the anti-CTLA-4 antibody is ipilimumab (also known as 10D1 , MDX-D010) a fully human monoclonal IgG antibody that binds to CTLA-4.
[0090] Other immune-checkpoint inhibitors include lymphocyte activation gene-3 (LAG-3) inhibitors, such as IMP321 , a soluble Ig fusion protein (Brignone et al., 2007, J. Immunol. 179:4202-421 1 ).
[0091] Other immune-checkpoint inhibitors include B7 inhibitors, such as B7-H3 and B7-H4 inhibitors. In particular, the anti-B7-H3 antibody MGA271 (Loo et al., 2012, Clin. Cancer Res. July 15 (18) 3834).
[0092] Also included are TIM3 (“T-cell immunoglobulin domain and mucin domain 3”) inhibitors (Fourcade et al., 2010, J. Exp. Med. 207:2175-86 and Sakuishi et al., 2010, J. Exp. Med. 207:2187- 94). The natural ligand of TIM-3 is galectin 9 (Gal9). Accordingly, the term “TIM-3 inhibitor” as used herein refers to a compound, substance or composition that can inhibit the function of TIM-3. For example, the inhibitor can inhibit the expression or activity of TIM-3, modulate or block the TIM-3 signaling pathway and/or block the binding of TIM-3 to galectin-9. Antibodies having specificity for TIM-3 are well known in the art and typically those described in WO201 1 155607, WO2013006490 and WO20101 17057.
[0093] In some embodiments, the immune checkpoint inhibitor is an Indoleamine 2,3-dioxygenase (IDO) inhibitor, preferably an IDO1 inhibitor. Examples of IDO inhibitors are described in WO 2014150677. Examples of IDO inhibitors include without limitation 1 -methyl-tryptophan (IMT), p- (3- benzofuranyl)-alanine, p-(3-benzo(b)thienyl)-alanine), 6-nitro-tryptophan, 6- fluoro-tryptophan, 4- methyl-tryptophan, 5 -methyl tryptophan, 6-methyl-tryptophan, 5-methoxy-tryptophan, 5 -hydroxy- tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-CI-indoxyl 1 ,3- diacetate, 9- vinylcarbazole, acemetacin, 5-bromo-tryptophan, 5-bromoindoxyl diacetate, 3- Amino- naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohydantoin derivative, a p-carboline derivative or a brassilexin derivative. Preferably the IDO inhibitor is selected from 1 -methyl-tryptophan, p-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3-Amino-naphtoic acid and p-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof.
[0094] In some embodiments, the immune checkpoint inhibitor is an anti-TIG IT (T cell immunoglobin and ITIM domain) antibody.
[0095] According to a specific embodiment, the anticancer treatment according to the present disclosure is an immune checkpoint inhibitor, preferably selected from an anti-PD-1 antibody and an anti-PD-L1 antibody.
[0096] In a preferred embodiment, the checkpoint blockade cancer immunotherapy agent is a PD-1 blocking antibody, such as Nivolumab or Pembrolizumab.
[0097] Compounds of formula (I)
[0098] Compounds of formula (I) can be synthesized by means of techniques that are routinely used by the skilled person.
[0099] According to a preferred embodiment, the compound of formula (I) is selected from the group consisting of:
[0100] N-(4-(3-((4-(Oct-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “BPR001 -615” (also called PB615 in the disclosure) of formula (II):
[0101] [Formula II] :
[0102] N-(4-(3-((4-(Hex-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB614”;
[0103] N-(4-(3-((4-(3-Hydroxyprop-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB608”;
[0104] N-(4-(3-((4-(Cyclohexylethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB612”;
[0105] N-(4-(3-((4-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB610”;
[0106] N-(4-(3-((4-((trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB671 ”;
[0107] N-(4-(3-((4-(dec-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB672”;
[0108] N-(4-(3-((4-(4-phenylbut-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB673”;
[0109] N-(4-(3-((3-(Oct-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB617”;
[0110] N-(4-(3-((3-(3-Hydroxyprop-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB619”;
[0111] N-(4-(3-((3-((Trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB620”;
[0112] N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide, herein referred to as “PB621 ”;
[0113] N-(3-(2-(Methylamino)thiazol-4-yl)phenyl)-4-(oct-1 -yn-1 -yl)benzenesulfonamide, herein referred to as “PB611”;
[0114] 4-(Oct-1 -yn-1 -yl)-N-(3-(2-(phenylamino)thiazol-4-yl)phenyl)benzenesulfonamide, herein referred to as “PB613”;
[0115] and
[0116] N-(3-(2-Aminothiazol-4-yl)phenyl)-4-(oct-1 -yn-1 -yl)benzenesulfonamide, herein referred to as “PB622”.
[0117] Methods for synthetizing these specific preferred compounds are disclosed in the experimental section below.
[0118] The compounds of formula (I) according to the present disclosure can be in the form of pharmaceutically acceptable salts. Pharmaceutically acceptable salts include the acid addition and base salts thereof.
[0119] Suitable acid addition salts are formed from acids, which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate,
nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate and xinafoate salts.
[0120] Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, 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. For a review on suitable salts, see "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley- VCH, Weinheim, Germany, 2002).
[0121] The compounds used in the context of the present disclosure may exist in both unsolvated and solvated forms. The term “solvate” describes a molecular complex comprising the benzene sulfonamide thiazole compound and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term “hydrate” is employed when said solvent is water. Also included are complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts. Also included are complexes of the drug containing two or more organic and/or inorganic components, which may be in stoichiometric or non-stoichiometric amounts. The resulting complexes may be ionised, partially ionized, or non-ionized. For a review of such complexes, see J Pharm Sci, 64 (8), 1269-1288 by Haleblian (August 1975).
[0122] The compounds of formula (I) thus include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof. The compounds of formula (I) include all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) and isotopically- labeled compounds of formula (I).
[0123] Therapeutic use
[0124] As mentioned above, the anticancer treatment and the compound of formula (I) according to the present disclosure, particularly a compound of formula (II), are used for the treatment of cancer in a patient.
[0125] The present disclosure thus provides a method for treating cancer comprising administering, to a patient in need thereof, a therapeutically effective amount of an anticancer treatment and of the compound of formula (I), particularly a compound of formula (II), as defined above.
[0126] The present disclosure further provides the compound of formula (I), particularly a compound of formula (II), for its use in the manufacture of a medicament for treating cancer, either as monotherapy or in the disclosed combinations.
[0127] According to the present disclosure, the anticancer treatment and the compound of formula (I), particularly a compound of formula (II), are administered to the patient either simultaneously, separately or sequentially in any order. According to a specific embodiment, the anticancer treatment is administered after the compound of formula (I), particularly a compound of formula (II). In other words, the anticancer treatment is administered to a patient who has already received the compound of formula (I), particularly a compound of formula (II). According to another specific embodiment, the
compound of formula (I), particularly a compound of formula (II), is administered after the anticancer treatment. In other words, the compound of formula (I) , particularly a compound of formula (II), is administered to a patient who has already received the anticancer treatment.
[0128] The terms “Subject” and “Patient” refer to a human or an animal suffering from cancer. Typically, the patient is a mammal. The patient can e.g. be a human, a feline such as a cat, a canine such as a dog or an equid such as a horse. Preferably, the patient is human.
[0129] For the avoidance of doubt, references herein to "treatment" include references to curative, palliative and prophylactic treatment. A “treatment” aims at reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or reversing, alleviating, inhibiting the progress of, or preventing one or more symptoms of the disorder or condition to which such term applies. In particular, in specific embodiments, when referring to cancer disorders, the term treatment may refer to slowing or preventing the growth of a tumor, or reducing the size of a tumor, or eradicating a tumor, or preventing metastatic development.
[0130] The term “cancer” herein refers to the physiological condition in subjects that is characterized by unregulated or dysregulated cell growth or death. The term "cancer" includes solid tumors and blood born tumors.
[0131] Typically, the combination for use according to the present disclosure applies to various organs of cancer origin (such as breast, colon, gastric, rectum, pancreatic, lung, skin, head and neck, bladder, ovary, prostate), and also to various cancer cell types (adenocarcinoma, squamous cell carcinoma, large cell cancer, melanoma, etc).
[0132] In a particular embodiment, the patient suffers from a solid cancer selected from the group consisting of skin cancer (e.g. melanoma, nonmelanoma skin cancer), colorectal cancer, adrenal cortical cancer, anal cancer, bile duct cancer (e.g. periphilar cancer, distal bile duct cancer, intrahepatic bile duct cancer), bladder cancer, bone cancer (e.g. osteoblastoma, osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma), sarcomas such as liposarcoma and soft-tissue sarcoma, brain and central nervous system cancer (e.g. meningioma, astocytoma, oligodendrogliomas, ependymoma, gliomas, medulloblastoma, ganglioglioma, germinoma, craniopharyngioma), breast cancer (e.g. ductal carcinoma in situ, infiltrating ductal carcinoma, infiltrating lobular carcinoma, lobular carcinoma in situ), cervical cancer, endometrial cancer (e.g. endometrial adenocarcinoma, adenocanthoma, papillary serous adnocarcinoma), esophagus cancer, gallbladder cancer (mucinous adenocarcinoma, small cell carcinoma), gastrointestinal carcinoid tumors (e.g. choriocarcinoma, chorioadenoma destruens), kidney cancer (e.g. renal cell cancer), laryngeal and hypopharyngeal cancer, liver cancer (e.g. hepatic adenoma, hepatocellular carcinoma), lung cancer (e.g. small cell lung cancer, non-small cell lung cancer), mesothelioma, nasal cavity and paranasal sinus cancer (e.g. esthesioneuroblastoma, midline granuloma), nasopharyngeal cancer, neuroblastoma, oral cavity and oropharyngeal cancer, ovarian cancer, pancreatic cancer, penile cancer, pituitary cancer, prostate cancer, retinoblastoma, rhabdomyosarcoma (e.g. embryonal rhabdomyosarcoma, alveolar rhabdomyosarcoma, pleomorphic rhabdomyosarcoma), salivary gland cancer, stomach cancer, testicular cancer (e.g. seminoma, nonseminoma germ cell cancer), thymus
cancer, thyroid cancer (e.g. follicular carcinoma, anaplastic carcinoma, poorly differentiated carcinoma, medullary thyroid carcinoma), vaginal cancer, vulvar cancer, and uterine cancer (e.g. uterine leiomyosarcoma).
[0133] In a particular embodiment, the patient suffers from a hematological cancer such as leukaemia, lymphoma (such as Hodgkin lymphoma or non-Hodgkin lymphoma) and myeloma.
[0134] As shown in the experimental section below, the compounds according to the present disclosure have been shown to have high potency against sensitive and resistant cancer cell lines from melanoma and colorectal cancer. Thus, according to another specific embodiment, the cancer treated according to the present disclosure is selected from skin cancer and colorectal cancer.
[0135] According to a preferred embodiment, the cancer treated according to the present disclosure is a colorectal cancer, a gastric cancer, a pancreatic cancer, a breast cancer, a lung cancer or skin cancer (preferably melanoma).
[0136] Without wishing to be bound by theory, the present inventors submit that the compounds of formula (I) described herein render the tumor immunogenic, i.e. detectable by the immune system, thereby allowing for the potentiation of the anticancer effect of the combined anticancer treatment.
[0137] Thus, according to a specific embodiment, the cancer according to the present disclosure is a non-immunogenic tumor. By “non-immunogenic tumor” it is herein referred to a tumor that does not elicit T-cell response. The skilled person is familiar with this notion and knows how to determine whether a tumor is immunogenic or not (see e.g. Wang et al. Elife 8 (2019): e49020). Such tumors are generally associated with:
[0138] - few tumor-infiltrating lymphocytes, and, accordingly with a low density of tumor-infiltrating lymphocytes, either at the margin or in the center of the tumor as e.g. determined in the international application published under reference W02007/045996 or according to the Immunoscore®;
[0139] - an exhausted population of CD8+ T cells - also referred to as “Tex cells” (see Wherry, E. John, and Makoto Kurachi, Nature Reviews Immunology 15.8 (2015): 486-499) which can be identified as shown in Bengsch et al. Immunity 48.5 (2018): 1029-1045;
[0140] - limited tumor antigen presentation (as shown in Wang (2019));
[0141] - a majority of M2 macrophages and myeloid-derived suppressor cells; and/or
[0142] - type-2 inflamed tumor microenvironment (see e.g. Gajewski et al. Tumor immune microenvironment in cancer progression and cancer therapy (2017): 19-31 or Trujillo et al. Cancer immunology research 6.9 (2018): 990-1000).
[0143] According to another embodiment, the cancer according to the present disclosure is “resistant” to immunotherapy meaning that the patient does not or poorly respond to immunotherapy, and particularly to an immune checkpoint inhibitor monotherapy.
[0144] As used herein, the term “responder” refers to a patient that will achieve a response, i.e. a patient where the cancer is eradicated, reduced or stabilized after treatment. A non-responder or
refractory patient includes patients for whom the cancer does not show reduction or stabilization after the immunotherapy, and particularly after the immune checkpoint therapy.
[0145] The present inventors have further showed that the combination therapy according to the present disclosure triggers remarkable tumor cell inhibition in cancer lines from tumors overexpressing GRP78.
[0146] Overexpression of GRP78 was shown to be associated with poor prognosis in multiple cancers such as ovarian cancer (see Samanta et al. Scientific reports 10.1 (2020): 1 -12), melanoma (Shimizu et al. Pathology & Oncology Research 23.1 (2017): 1 1 1 -1 16), lung cancer (Xia et al. Journal of Translational Medicine 19.1 (2021 ): 1 -14), gastric cancer (Zhang et al. Clinical & experimental metastasis 23.7 (2006): 401 -410), breast cancer (Oncology letters 10.4 (2015): 2149-2155., esophageal cancer (Zhao et al. Digestive diseases and sciences 60.9 (2015): 2690-2699), pancreatic cancer (Tong et al. Pancreatology 21 .7 (2021 ): 1378-1385) or colorectal cancer (Thornton et al. International journal of cancer 133.6 (2013): 1408-1418). The combination according to the present disclosure allows significantly reducing GRP78 levels.
[0147] The combination therapy according to the present disclosure can thus be particularly beneficial to patients presenting high levels of GRP78 and who are therefore identified as having a poor prognosis.
[0148] Therefore, according to a further embodiment, the patient according to the present disclosure overexpresses GRP78.
[0149] “78 kDa glucose-regulated protein” or “GRP78”, also known as “Binding immunoglobulin protein” (BiP) or “heat shock 70 kDa protein 5” (HSPA5) is a protein that in humans is encoded by the HSPA5 gene. It is an endoplasmic reticulum chaperone that plays a key role in protein folding and quality control in the endoplasmic reticulum lumen. The sequence of GRP78 is available under reference P1 1021 in the Uniprot database library.
[0150] By “overexpresses” it is meant that the level of GRP78 measured in a biological sample obtained from the patient is significantly higher than the level measured in a control sample. Typically, said control sample can be a sample obtained from a control population or from a control tissue. A control population is typically constituted of healthy subjects, i.e. subjects who do not suffer from cancer or any other disease. A control tissue is typically constituted of healthy tissues, i.e. tissues not affected by a disease. Said control tissue is usually obtained from the patient himself. The skilled person knows several techniques that allow determining whether a gene/protein is overexpressed as compared to a control sample. The skilled person is familiar with such techniques which are used routinely. Several studies have been published with respect to the evaluation of GRP78 levels (see e.g. Huang et al (2018). International Journal of Clinical and Experimental Pathology, 77(1 1 ), 5223). Typically, the level of GRP78 is considered as “overexpressed” when said level is 1 .5, preferably 1 .7, more preferably 2 times higher than the level measured in a control population.
[0151] Typically, the GRP78 level measured is either the circulating GRP78 level or the intratumor GRP78 level.
[0152] “Circulating GRP78” refers to the GRP78 protein present in the blood circulation of the patient, typically in the cell-free section of the blood (plasma/serum) of the patient. The level of circulating GRP78 protein is typically measured by evaluating the quantity of GRP78 in a blood sample, typically a plasma/serum sample obtained from the patient. The control used for determining whether a level of circulating GRP78 is overexpressed is typically the GRP78 level measured in blood samples from a control population.
[0153] “Intratumor GRP78” refers to the GRP78 protein expressed within the tumor of the patient. The level of the GRP78 protein is typically measured by evaluating the quantity of GRP78 expressed in a tumor sample, typically in a tumor biopsy obtained from the patient. The control used for determining whether the level of intratumor GRP78 is overexpressed is typically the GRP78 level measured in control tissues, i.e. healthy tissues from the patient.
[0154] The person skilled in the art is familiar with many techniques that are used on a daily basis to determine the expression level of a protein such as GRP78.
[0155] Such methods typically involve contacting the biological sample to be analyzed with an agent capable of specifically binding the target protein. This agent is usually a polyclonal or monoclonal antibody. The presence of the protein is then typically detected by standard immunodetection methods after separation of the proteins by electrophoresis (technique also called "Western blotting") or by immunoassays by direct, indirect, competition or immunocapture methods (techniques also called "ELISA"). The formation of a complex between the protein of interest and the antibody(s) targeting said protein is usually detected and quantified by measuring an enzymatic reaction generating a colored, chemiluminescent or fluorescent product leading in a specific staining pattern with a staining %. Typically “strong staining” is defined as >50% of the tumor cells staining positive, “moderate staining” is defined as 10 to <50% of the tumor cells staining positive, and “weak staining” is defined as <10% of the tumor cells staining positive (Samanta et al, 2020).
[0156] Several kits are currently available for measuring the plasma/serum level GRP78 level. For instance, the ELISA kit commercialized by Enzo Life Sciences under reference ADI-900-214 can be used.
[0157] It is also possible to determine the intratumor GRP78 level by determining the density of cells expressing GRP78. Typically, methods for measuring the density GRP78 expressing cells comprise a step of contacting the tumor tissue sample with at least one selective binding agent capable of selectively interacting with GRP78. The selective binding agent may be a polyclonal antibody or a monoclonal antibody, an antibody fragment, synthetic antibodies, or other protein-specific agents such as nucleic acid or peptide aptamers. The skilled person knows several antibodies which are specific to GRP78. Many of these antibodies are commercially available. Immunohistochemistry is particularly suitable for evaluating the density of GRP78 cells. Typically, the tissue tumor sample is firstly incubated with labelled antibodies directed against GRP78. After washing, the labelled antibodies which are bound to GRP78 are revealed by the appropriate technique, depending of the kind of label born by the labelled antibody, e.g. radioactive, fluorescent or enzyme label.
[0158] The level of GRP78 can also be measured by determining the quantity of mRNA produced by the HSPA5 gene. Methods for determining a quantity of mRNA are well known in the art. For example, nucleic acid contained in the samples is first extracted according to standard methods, for example using lytic enzymes or chemical solutions or extracted by nucleic-acid-binding resins following the manufacturer's instructions. The extracted mRNA is then detected by hybridization (e. g., Northern blot analysis) and/or amplification (e.g., RT-PCR). Quantitative or semi-quantitative RT- PCR are preferred methods.
[0159] Accordingly, according to a specific embodiment, the present disclosure provides a method for treating cancer in a patient comprising a step of measuring the level of GRP78 in a tumor or blood/plasma/serum sample obtained from said patient, and then a step of administering a therapeutically effective amount of an anticancer treatment and of a compound of formula (I), particularly a compound of formula (II), as defined above if said patient is identified as overexpressing GRP78.
[0160] The compounds used in the context of the present disclosure may be administered by any suitable route. The skilled person knows which route of administration to use as well as the corresponding dosages. Compounds useful in the context of the present disclosure are typically administered via parenteral (e.g., intravenous, intramuscular or subcutaneous) or via oral administration.
[0161] Another aspect of the disclosure is a compound of Formula (I):
Formula (I) wherein
Ri represents H, methyl, phenyl or acetyl;
R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4, for use in the treatment of a GRP78 overexpressing tumor in a patient in need thereof.
[0162] In a specific embodiment, a “GRP78 overexpressing tumor” means a tumor from a patient characterized by a positive staining of GRP78 for at least 1 %, preferably at least 10% of the tumor cells in a tumor sample. Such staining may be measured by methods well known in the art and particularly as described in Samanta et al., 2020.
[0163] In an embodiment, a “GRP78 overexpressing tumor” may be characterized by measuring GRP78 expression at intratumor or circulating level as described above.
[0164] The compounds of formula (I), particularly the compound of formula (II), either in monotherapy or in combination treatment as disclosed herein, is useful for patients having GRP78 overepressing tumor, whether high GRP78 tumor or low GRP78 tumor.
[0165] In one embodiment, the compound of formula I or formula II is for use in the treatment of a GRP78 positive tumor in patient in need thereof, wherein said tumor is a high GRP78 tumor.
[0166] By “high GRP78 tumor”, it is meant a tumor from a patient characterized by a positive staining of GRP78 for at least 50% of the tumor cells present in a sample obtained from said tumor.
[0167] In one embodiment, the compound of formula I or formula II is for use in the treatment of a GRP78 positive tumor in patient in need thereof, wherein said tumor is a low GRP78 tumor.
[0168] By “low GRP78 tumor”, it is meant a tumor from a patient characterized by a positive staining of GRP78 for 1 % to less than 50%, preferably 10% to less than 50%, of the tumor cells present in a sample obtained from said tumor.
[0169] In a particular embodiment, the compound of formula I is for use in the treatment of a GRP78 overexpressing tumor in patient in need thereof, wherein said patient has a low GRP78 tumor and wherein the compound of formula (I) is the compound BPR001 -615 of formula (II) described above.
[0170] In one embodiment, the patient to be treated with the compound of formula I, particularly compound BPR001 -615 of formula (II) is having a low GRP78 tumor, i.e. a patient tumor with GRP78 staining by immunohistochemistry shows a staining with 1 % to less than 50%, preferably 10% to less than 50%, of positive tumor cells, as determined according to Samanta et al. 2020, meaning that between 1 % to less than 50%, preferably 10% to less than 50%, of the tumor cells staining is positive, as described in Samanta et al, 2020.
[0171] In one embodiment, the patient to be treated with the combination is having a high GRP78 tumor, i.e. a patient tumor with GRP78 staining by immunohistochemistry shows a staining with at least 50% of positive tumor cells, as determined according to Samanta et al. 2020, meaning that between 50% to less than 100% of the tumor cells staining is positive, as described in Samanta et al, 2020.
[0172] The compound of formula (I), particularly a compound of formula (II), as described herein is for use in the treatment of a GRP78 positive tumor, either a low GRP78 tumor or a high GRP78 tumor, in a patient in need thereof either alone as monotherapy or in combination with an anticancer agent, such combination being described in detail in the present disclosure.
[0173] The disclosure will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
Examples
[0174] Synthesis of the compounds of formula (I):
[0175] Chemical synthesis and characterization
[0176] Methanol, ethanol, pyridine, DMF, ethyl acetate, diethyl ether and dichloromethane were purchased from Sigma Aldrich. DMF was dried by distillation under reduced pressure over MgSCk, pyridine and triethylamine were distilled over CaH2 under reduced pressure while methanol, ethanol, diethyl ether, ethyl acetate and dichloromethane were used as received. All chemicals were purchased from Aldrich, Merck or Alfa Aesar and used without further purification. Thin layer chromatography (TLC) was performed on precoated Merck 60 GF254 silica gel plates and revealed first by visualization under UV light (254 nm and 365 nm) then spraying (ninhydrin or F SO EtOH). 1H and 13C NMR spectra were recorded on a Bruker Advance 200 MHz spectrometer or a Bruker Advance 400 MHz. Mass spectra (ESI-MS) were recorded on a Bruker (Daltonics Esquire 3000+). HRMS spectra were recorded on a ThermoFisher Q Exactive (ESI-MS) at a resolution of 140 000 at m/z 200. The purity of compounds was further assayed by HPLC analysis on a JASCO PU-2089 apparatus with Supelco analytical column Ascentis Express C18, 100mm x 46 mm 5 pM. UV- detection: 214; 254; 280; 360 nm. Eluent A: water with 1%0 formic acid, Eluent B: CH3CN with 1%0 formic acid. Method 1 : 0-1 min: 0%B; 1 -10 min: 0-100%B; 10-15 min: 100%B. Method 2: 0-2 min: 10%B; 2-9 min: 10-100%B; 9-18 min: 100%B. Method 3: 0-1 min: 30%B; 1 -6 min: 30-100%B; 6-8.5 min: 100%B.
[0177] General procedure for the Sonogashira coupling
[0178] To a suspension of haloaryle derivative (5a: N-(4-(3-((4- bromophenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide or 6: N-(4-(3-((3- bromophenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide) (1 eq.) in a mixture of EtsN:benzene (1/1 , [0.17M]) under argon were added Pd(PPhs)4 (15% mol.), Copper (I) iodide (15% mol.) and corresponding alkyne (5eq.). The resulting mixture was stirred at 80°C until complete conversion of the starting material (2h approx.). The reaction mixture was then cooled to room temperature and all volatiles were removed under reduced pressure and the crude material was purified by silica gel flash chromatography to afford the pure corresponding coupling product.
[0179] Synthetic procedures and characterizations
[0180] N-(4-(3-((4-(Oct-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (BPR001-615). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (400.0 mg, 0.88 mmol), Pd(PPhs)4 (152.5 mg, 15% mol.), copper (I) iodide (25.1 mg, 15%mol) and oct-1 -yne (653.2 pL, 4.43 mmol). Purification by silica gel flash chromatography (CH2Cl2:EtOAc; 100:0 to 70:30) afforded alkyne 1 as a white-yellowish powder (330.1 mg, 78%). 1H NMR (CD3OD, 200 MHz): <50.97 - 0.78 (m, 3H, H23), 1 .62 - 1 .15 (m, 9H, H22, H21, H20 and H19), 2.19 (s, 3H, Hi), 2.34 (t, J = 6.8 Hz, 2H, His), 6.97 (ddd, J = 8.0, 2.1 and 0.9 Hz, 1 H, H9), 7.26 - 7.14 (m, 2H, H4 and H10), 7.38 (d, J = 8.5 Hz, 2H, H14), 7.56 (d, J = 7.9 Hz, 1 H, Hu), 7.74 - 7.65 (m, 3H, H7 and H13). 13C NMR (CD3OD, 50 MHz): <514.5, 20.2, 22.7, 23.7, 29.7, 29.8, 32.6, 80.4, 95.4, 109.3, 120.1 , 121 .7, 123.6, 128.4 (2C), 130.3, 130.5, 132.9 (2C), 137.1 , 139.3, 139.7, 150.4, 159.6, 171 .0. HRMS-ESI (m/z) [M+H]+ calcd for C25H28N3O3S2, 482.1567; Found: 482.1565. HPLC (A280): Purity 97.8%; fa: 8.86 min (method 1 ).
[0181] N-(4-(3-((4-(Hex-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB614). To a solution of bromoaryle 5a (150.0 mg, 0.33 mmol) in a mixture of triethylamine (1 mL) and benzene
(1 mL) under argon, was added Pd(PPhs)4 (57.8 mg, 15% mol.) and copper (I) iodide (9.5 mg, 15% mol.). n-Hex-1 -yne (188.0 pL, 1 .66 mmol) was added to the medium and the resulting mixture was stirred for 2h at 80°C (TLC monitoring). The reaction mixture was cooled to r.t. and all volatiles were removed under reduced pressure. Purification by silica gel column chromatography (CH2Cl2:EtOAc; 100:0 to 70:30) afforded alkyne 7 as an off-white powder (1 17.2 mg, 78%). 1H NMR (CD3OD, 200 MHz): <5 0.91 (t, J = 7.1 Hz, 3H, H21), 1 .62 - 1 .35 (m, 4H, H20 and H19), 2.20 (s, 3H, H 1 ), 2.37 (t, J = 6.8 Hz, 2H, His), 6.97 (ddd, J = 8.0, 2.2, 0.9 Hz, 1 H, H9), 7.27 - 7.16 (m, 2H, H4 and H10), 7.39 (d, J = 8.5 Hz, 1 H, Hu), 7.57 (dt, J = 7.8, 1 .2 Hz, 1 H, Hu), 7.76 - 7.64 (m, 2H, H7 and H13). 13C NMR (CD3OD, 50 MHz): <5 14.1 , 19.8, 22.7, 23.1 , 31 .9, 80.4, 95.4, 109.3, 120.2, 121 .8, 123.6, 128.4 (2C), 130.4, 130.5, 132.9 (2C), 137.2, 139.3, 139.7, 150.5, 159.6, 171 .0. HRMS-ESI (m/z): [M+H]+ calcd for C23H24N3O3S2, 454.1254; Found: 454.1249. HPLC (A280): Purity >99%; fa: 8.08 min (method 1 ).
[0182] N-(4-(3-((4-(3-Hydroxyprop-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB608). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (300.0 mg, 0.66 mmol), Pd(PPhs)4 (1 14.4 mg, 15% mol.), copper (I) iodide (18.9 mg, 15% mol.) and propargyl alcohol (190.4 pL, 3.30 mmol). Purification by silica gel flash chromatography (CH2Cl2:CH3OH; 100:0 to 95:5) afforded alkyne 8 as a white-yellowish powder (201 .9 mg, 72%). 1H NMR (CD3OD, 200 MHz): <5 2.20 (s, 1 H, Hi), 4.36 (s, 2H, His), 6.97 (ddd, J = 8.0, 2.2, 1 .0 Hz, 1 H, H9), 7.34 - 7.14 (m, 2H, H4 and H10), 7.51 - 7.42 (m, 2H, H14), 7.63 - 7.53 (m, 1 H, Hu), 7.78 - 7.66 (m, 3H, H7 and H13). 13C NMR (CD3OD, 50 MHz): <522.7, 51 .2, 84.0, 92.7, 109.3, 120.2, 121 .8, 123.7, 128.5 (2C), 129.1 , 130.6, 133.1 (2C), 137.2, 139.3, 140.6, 150.5, 159.6, 171 .1 . HRMS-ESI (m/z): [M+H]+ calcd for C20H18N3O4S2, 428.0733; Found: 428.0736. HPLC (A254): Purity 99.1 %; fa: 5.76 min (method 2).
[0183] N-(4-(3-((4-(Cyclohexylethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB612). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (1 10 mg, 0.24 mmol), Pd(PPhs)2Cl2 (25.7 mg, 15% mol.), copper (I) iodide (7.0 mg, 15% mol.) and cyclohexylacetylene (159.4 pL, 1 .22 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 70:30) afforded alkyne 9 as a white-yellowish powder (55 mg, 47%). 1H NMR (acetone-d6, 200 MHz): <5 1 .58 - 1 .22 (m, 6H, H20 and H21), 1 .91 - 1 .61 (m, 4H, H19), 2.28 (s, 3H, Hi), 2.70 - 2.49 (m, 1 H, His), 7.09 (ddd, J = 8.0, 2.3, 1 .1 Hz, 1 H, H9), 7.26 (t, J = 7.9 Hz, 1 H, H10), 7.38 (s, 1 H, H4), 7.48 (d, J = 8.4 Hz, 2H, H14), 7.61 (d, J = 7.7 Hz, 1 H, Hu), 7.75 (d, J = 8.5 Hz, 2H, H13), 7.85 (t, J = 1 .9 HZ, 1 H, H7), 9.10 (S, 1 H, NHsulfonamide), 1 1 .17 (S, 1 H, NHacetyl). 13C NMR (acetone-d6, 50 MHz): <522.9, 25.4 (2C), 26.5, 30.3, 33.2 (2C), 80.3, 99.0, 108.8, 1 19.6, 121 .2, 123.1 , 128.2 (2C), 129.7, 130.3, 132.7 (2C), 136.8, 139.0, 139.6, 149.7, 159.0, 169.2. HRMS-ESI (m/z): [M+H]+ calcd for C25H26N3O3S2, 480.1410; Found: 480.1413. HPLC (A254): Purity 98.9%; fa: 6.23 min (method 2).
[0184] N-(4-(3-((4-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB610). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (300.0 mg, 0.66 mmol), Pd(PPhs)4 (1 14.4 mg, 15% mol.), copper (I) iodide (18.9 mg, 15% mol.) and trimethylsilylacetylene (281 .9 pL, 1 .98 mmol). After completion of the reaction (about 2h), the mixture was filtered through
a pad of silica (eluted with CH2Cl2/EtOAc 70:30) and the filtrate was concentrated under reduced pressure. The black residue was dissolved in CH2CI2/CH3OH (1 :1 ; 2mL:2mL) and K2CO3 (456.1 mg, 3.3 mmol) was added to the mixture. The resulting suspension was stirred at r.t. for 5h., adsorbed onto silica prior to purification by silica gel flash chromatography (CH2CI2/CH3OH; 100:0 to 95:0) to afford the alkyne 11 as a yellow-brownish powder (88.2 mg, 34%). 1H NMR (CD3OD, 200 MHz): <5 2.20 (s, 3H, Hi), 3.70 (s, 1 H, H17), 6.97 (ddd, J = 7.9, 2.3, 1 .1 Hz, 1 H, H9), 7.30 - 7.14 (m, 2H, H10 and H4), 7.62 - 7.44 (m, 3H, Hu and H14), 7.80 - 7.67 (m, 3H, H7 and H13). 13C NMR (CD3OD, 50 MHz): <5 22.7, 82.4, 82.9, 109.4, 120.2, 121 .8, 123.7, 128.4 (3C), 130.6, 133.6 (2C), 137.2, 139.2, 141 .0, 150.4, 159.6, 171 .0. HRMS-ESI (m/z): [M+H]+ calcd for C19H16N3O3S2, 398.0628; Found: 298.0626. HPLC (A254): Purity 98.9%; fa: 6.23 min (method 2).
[0185] N-(4-(3-((4-((trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB671). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (22.0 mg, 7.5% mol.), copper (I) iodide (5.0 mg, 10% mol.) and trimethylsilylacetylene (178.0 pL, 1 .25 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 50:50) afforded alkyne 10 as an off-white solid powder (100.0 mg, 85%). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.60. 1H NMR (Acetone-cfe, 400 MHz): <5 0.21 (s, 9H, H20), 2.28 (s, 3H, Hi), 7.08 (ddd, J = 8.0, J = 2.3, J = 1 .1 Hz, 1 H, Haro), 7.26 (t, J = 7.9 Hz, 1 H, Haro), 7.38 (s, 1 H, H4), 7.54 - 7.61 (m, 2H, Haro), 7.62 (ddd, J = 7.8, J = 1 .7, J = 1 .0 Hz, 1 H, Haro), 7.75 - 7.82 (m, 2H, Haro), 7.86 (t, J= 1 .9 Hz, 1 H, Haro), 9.10 (s, 1 H, NHsulfonamide), 1 1 .14 (s, 1 H, NHacetyi). 13C NMR (Acetone-cfe, 101 MHz): <50.3 (3C), 22.8, 98.6, 104.0, 108.8, 1 19.60, 121 .3, 123.1 , 128.2 (2C), 128.3, 130.3, 133.0, 136.7, 138.8, 140.5, 149.6, 159.0, 169.1 . HRMS-ESI (m/z): [M+H]+ calcd for C22H24N3O3S2SI, 470.10229; Found: 470.10260. HPLC (A254): Purity 99.8%; fa: 10.71 min (method 3).
[0186] N-(4-(3-((4-(dec-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB672). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (339.0 mg, 0.75 mmol), Pd(PPhs)4 (66.0 mg, 7.5% mol.), copper (I) iodide (15.0 mg, 10% mol.) and dec-1 -yne (677.0 pL, 3.75 mmol). Purification by silica gel flash chromatography (petroleum ether:EtOAc; 100:0 to 50:50) afforded alkyne 12 as an off-white solid powder (323.5 mg, 85%). f (EtOAc/cyclohexane, 1/1, v/v) = 0.65. 1 H NMR (Acetone-d6, 400 MHz): 5 0.81 - 0.88 (m, 3H, H27), 1 .23 - 1 .32 (m, 8H, Haiiph), 1 .41 (t, J = 7.3 Hz, 2H, Haiiph), 1 .50 - 1 .59 (m, 2H, Haiiph), 2.28 (s, 3H, Hi ), 2.40 (t, J = 7.0 Hz, 2H, H20), 7.09 (ddd, J= 8.0, J = 2.2, J= 1 .0 Hz, 1 H, Haro), 7.26 (t, J= 7.9 Hz, 1 H, Haro), 7.37 (s, 1 H, Haro), 7.45 - 7.53 (m, 2H, Haro), 7.61 (dt, J = 7.8, J = 1 .3 Hz, 1 H, Haro), 7.73 - 7.79 (m, 2H, Haro), 7.86 (t, J = 1 .9 Hz, 1 H, Haro), 9.06 (s, 1 H, NHsulfonamide), 1 1 .15 (s, 1 H, NHacetyi). 13C NMR (Acetone-d6, 101 MHz): <5 14.3, 19.7, 22.8, 23.3, 29.2, 29.5, 29.7, 29.8, 29.9, 32.5, 95.2, 108.8, 1 19.5, 121 .2, 123.0, 128.1 (2C), 129.6, 130.2, 132.6 (2C), 136.7, 138.9, 139.6, 149.6, 159.0, 169.1 . HRMS-ESI (m/z) [M+H]+ calcd for C27H32N3O3S2: 510.18796; Found: 510.18829. HPLC (A254): Purity 97.2%; fa: 10.42 min (method 3).
[0187] N-(4-(3-((4-(4-phenylbut-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB673). The general procedure for Sonogashira coupling was followed using bromoaryle 5a (339.0
mg, 0.75 mmol), Pd(PPhs)4 (66.0 mg, 7.5% mol.), copper (I) iodide (15.0 mg, 10% mol.) and dec-1 - yne (677.0 pL, 3.75 mmol). Purification by silica gel flash chromatography (petroleum ether:EtOAc; 100:0 to 50:50) afforded alkyne 13 as an off-white solid powder (305.8 mg, 81 %). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.48. 1H NMR (400 MHz, Acetone-cfe) 6 2.28 (s, 3H, Hi), 2.70 (t, J = 7.2 Hz, 2H, H20), 2.87 (t, J = 7.4 Hz, 2H, H21), 7.09 (ddd, J = 8.1 , J = 2.3, J = 1 .0 Hz, 1 H, Haro), 7.19 (dq, J = 7.8, J = 2.8 Hz, 1 H, Haro), 7.22 - 7.33 (m, 5H, Haro), 7.37 (s, 1 H, H4), 7.40 - 7.49 (m, 2H, Hara), 7.61 (dt, J = 7.9, J = 1 .3 Hz, 1 H, Haro), 7.72 - 7.78 (m, 2H, Haro), 7.86 (t, J = 1 .9 Hz, 1 H, Haro), 9.10 (s, 1 H, NHsuifonamide), 1 1 .16 (s, 1 H, NHacetyi). 13C NMR (Acetone-d6, 101 MHz): <522.0, 22.8, 35.5,
80.7, 94.5, 108.8, 1 19.5, 121 .1 , 123.0, 127.1 , 128.1 (2C), 129.1 (2C), 129.4, 129.4 (2C), 130.2, 132.6 (2C), 136.7, 138.9, 139.6, 141 .4, 149.6, 159.0, 169.1 . HRMS-ESI (m/z): [M+H]+ calcd for C27H24N3O3S2: 502.12536; Found: 502.12558. HPLC (A254): Purity 99.8%; fa: 10.43 min (method 3).
[0188] N-(4-(3-((3-(Oct- 1 -yn- 1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB617). The general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and oct-1 -yne (185.0 pL, 1 .25 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 60:40) afforded alkyne 14 as a white-yellowish powder (103.0 mg, 86%). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.57. 1H NMR (CDCI3, 200 MHz): <50.86 (t, J = 6.0 Hz, 3H, H25), 1 .41 - 1 .20 (m, 6H, H22, H23 and H24), 1 .63 - 1 .45 (m, 2H, H21), 2.04 (s, 1 H, Hi), 2.33 (t, J = 6.9 Hz, 2H, H20), 7.00 - 6.85 (m, 2H, Haro), 7.12 (t, J = 8.0 Hz, 1 H, H10), 7.34 - 7.21 (m, 2H, Haro), 7.50 - 7.39 (m, 3H, Haro), 7.61 (d, J = 7.9 Hz, 2H, Haro), 7.83 (s, 1 H, Haro), 10.51 (s, 1 H, NHsuifonamide). 13C NMR (CDCI3, 50 MHz): <5 14.3, 19.6, 22.7, 23.2,
28.7, 28.8, 31 .5, 79.1 , 93.6, 108.9, 120.3, 121 .7, 123.5, 125.8, 126.0, 129.2, 129.9, 130.3, 135.7, 136.1 , 136.9, 139.5, 148.7, 159.0, 168.6. HRMS-ESI (m/z) [M+H]+calcd for C25H28N3O3S2, 482.1567; Found: 482.1574. HPLC (A254): Purity 98.9%; fa: 8.92 min (method 1 ).
[0189] N-(4-(3-((3-(3-Hydroxyprop-1-yn-1-yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB619). The general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and propargyl alcohol (72.0 pL, 1 .25 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 60:40) afforded alkyne 15 as a white powder (76.1 mg, 71 %), along with unreacted starting material 6 (22.0 mg, 20%). Rf (EtOAc/cyclohexane, 2/1, v/v) = 0.38. 1H NMR (CD3OD, 200 MHz): <5 2.21 (s, 3H, Hi), 4.37 (s, 2H, H20), 6.99 (ddd, J = 8.1 , 2.3, 1 .1 Hz, 1 H, H9), 7.29 - 7.17 (m, 2H, H4 and H10), 7.41 (t, J = 7.8 Hz, 1 H, Hi 6) , 7.62 - 7.51 (m, 2H, Haro), 7.71 (tt, J = 3.4, 1 .5 Hz, 2H, Haro), 7.80 (t, J = 1 .8 Hz, 1 H, Haro). 13C NMR (CD3OD, 50 MHz): <5 22.7, 51 .2, 83.7, 91 .2, 109.3, 120.1 ,
121 .7, 123.7, 125.5, 128.0, 130.4, 130.6, 131 .0, 136.7, 137.2, 139.2, 141 .6, 150.4, 159.6, 171 .1 . HRMS-ESI (m/z) [M+H]+ calcd for C20H18N3O4S2, 428.0733; Found: 428.0737. HPLC (A254): Purity 97.5%; fa: 10.1 1 min (method 3).
[0190] N-(4-(3-((3-((Trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB620). The general procedure for Sonogashira coupling was followed using bromoaryle 6 (1 13.0 mg, 0.25 mmol), Pd(PPhs)4 (44.0 mg, 15% mol.), copper (I) iodide (5.0 mg, 10% mol.) and trimethylsilylacetylene (178.0 pL, 1 .25 mmol). Purification by silica gel flash chromatography
(cyclohexane:EtOAc; 100:0 to 50:50) afforded alkyne 16 as a pale yellow powder (98.2 mg, 84%). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.52. 1H NMR (CDCh, 200 MHz): <5 0.17 (s, 9H, H20), 1 .94 (s, 3H, Hi), 6.89 (d, J = 7.1 Hz, 2H, Haro), 7.08 (t, J = 7.8 Hz, 1 H, Haro), 7.46 - 7.17 (m, 3H, Haro), 7.53 (d, J = 7.5 Hz, 1 H, Haro), 7.66 (d, J = 8.0 Hz, 1 H, Haro), 7.90 (s , 1 H, Haro), 8.18 (S, 1 H, NHsulfonamide), 10.97 (s, 1 H, NHacetyi). 13C NMR (CDCI3, 50 MHz): <50.1 (3C), 23.0, 97.5, 102.9, 109.0, 120.0, 121 .5, 123.4, 124.7, 126.9, 129.3, 129.9, 130.6, 135.6, 136.4, 136.9, 139.6, 148.6, 159.4, 169.0. HRMS-ESI (m/z): [M+H]+ calcd for C22H24N3O3S2Si, 470.1023; Found: 470.1033. HPLC (A254): Purity 96.6%; fa: 10.62 min (method 3).
[0191] N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide (PB621). To a yellow solution of alkyne 16 (51 mg, 0.108 mmol) in methanol (4 mL) was added solid K2CO3 (138 mg, 1 .0 mmol). The resulting suspension was stirred at r.t. for 14h. The mixture was partitioned between EtOAc and water, the aqueous phase was extracted with EtOAc once. The combined organic layers were washed with brine, dried with Na2SO4 and concentrated under reduced pressure. Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 50:50) afforded alkyne 17 as a white powder (41 .8 mg, 97%). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.47. 1H NMR (CD3OD, 200 MHz): <52.21 (s, 3H, H 1 ), 3.64 (s, 1 H, Haikyne), 6.99 (ddd, J = 8.1 , 2.3, 1 .1 Hz, 1 H, H9), 7.33 - 7.16 (m, 2H, H4 and H10), 7.42 (t, J = 7.8 Hz, 1 H, HI6), 7.59 (ddt, J = 7.8, 3.5, 1 .4 Hz, 2H, Haro), 7.73 (dt, J = 6.5, 1 .6 Hz, 2H, Haro), 7.85 (t, J= 1 .7 Hz, 1 H, Haro). 13C NMR (CD3OD, 50 MHz): <522.7, 81 .1 , 82.7, 109.3, 120.2, 121 .7, 123.7, 124.9, 128.4, 130.4, 130.6, 131 .5, 137.2, 137.2, 139.2, 141 .6, 150.4, 159.6, 171.1 . HRMS-ESI (m/z): [M+H]+ calcd for Ci9Hi6N3O3S2, 398.0628; Found: 398.0626. HPLC (A254): Purity 96.9%; fa: 10.84 min (method 3).
[0192] N-(3-(2-(Methylamino)thiazol-4-yl)phenyl)-4-(oct- 1 -yn- 1 -yl)benzenesulfonamide (PB611 ). The Sonogashira coupling was performed using 5b (325.0 mg, 0.77 mmol), Pd(PPh3)2CI2 (80.6 mg, 15% mol.), copper (I) iodide (21 .9 mg, 15% mol.) and oct-1 -yne (565.0 pL, 3.83 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 70:30) afforded 21 as an off-white powder (80.1 mg, 23%). 1H NMR (Acetone-d6, 200 MHz): <5 0.85 (t, J = 6.4 Hz, 3H, H22), 1 .70 - 1 .18 (m, 8H, His, H19, H20 and H2I), 2.40 (t, J = 6.8 Hz, 2H, H17), 3.12 - 2.88 (m, 3H, Hi), 6.73 (s, 1 H, NHthiazoie), 6.87 (s, 1 H, H3), 7.1 1 (ddd, J = 8.0, 2.2, 1 .2 Hz, 1 H, He), 7.23 (t, J = 7.8 Hz, 1 H, H9), 7.53 - 7.44 (m, 2H, H13), 7.58 (dt, J = 7.7, 1 .4 Hz, 1 H, H10), 7.91 - 7.70 (m, 3H, H6 and H <2), 9.10 (s, 1 H, NHsulfonamide). 13C NMR (Acetone-d6, 50 MHz) : <5 14.4, 19.8, 23.2, 29.2, 29.3, 31 .8, 32.1 , 80.2, 95.2, 102.0, 1 19.5, 120.6, 123.0, 128.2 (2C), 129.6, 130.0, 132.7 (2C), 137.4, 138.8, 139.7, 151 .4, 170.7. HRMS-ESI (m/z): [M+H]+ calcd for C24H23N3O2S2, 454.1618; Found: 454.1615. HPLC (A254): Purity 96.6%; fa: 12.03 min (method 3).
[0193] 4-(Oct-1-yn-1-yl)-N-(3-(2-(phenylamino)thiazol-4-yl)phenyl)benzenesulfonamide (PB613). The Sonogashira coupling was performed using 5c (100.0 mg, 0.21 mmol), Pd(PPh3)2CI2 (21 .6 mg, 15% mol.), copper (I) iodide (5.9 mg, 15% mol.) and oct-1 -yne (151 .7 pL, 1 .03 mmol). Purification by silica gel flash chromatography (cyclohexane:EtOAc; 100:0 to 70:30) afforded 22 as a brown powder (72 mg, 65%). 1H NMR (Acetone-d6, 200 MHz): <5 0.86 (t, J = 6.2 Hz, 3H, H25), 1 .67 - 1 .1 1 (m, 8H, H2I , H22, H23 and H24), 2.39 (t, J = 6.7 Hz, 2H, H20), 7.01 (t, J = 7.3 Hz, 1 H, HI2), 7.13 (d, J = 9.1 Hz,
2H, H6 and Hu), 7.33 (dt, J = 15.6, 7.8 Hz, 3H, Hi and H3), 7.48 (d, J = 8.2 Hz, 2H, HI6), 7.66 (d, J = 7.7 Hz, 1 H, H13), 7.88 - 7.73 (m, 4H, H2 and H15), 7.92 (t, J= 1 .9 Hz, 1 H, H9), 9.14 (s, 1 H, NHthiazoie), 9.35 (s, 1 H, NHsuifonamide). 13C NMR (Acetone-cfe, 50 MHz): <5 14.4, 19.8, 23.2, 29.2, 29.3, 32.1 , 80.2, 95.3, 103.8, 1 18.3 (2C), 1 19.5, 120.8, 122.6, 123.0, 128.2 (2C), 129.7, 129.9 (2C), 130.3, 132.7 (2C), 137.0, 139.0, 139.7, 142.4, 151 .3, 164.5. HRMS-ESI (m/z) [M+H]+calcd for C29H30N3O2S2, 516.1774; Found: 516.1777. HPLC (A280): Purity 97.3%; fa: 1 1 .96 min (method 3).
[0194] N-(3-(2-Aminothiazol-4-yl)phenyl)-4-(oct-1-yn-1-yl)benzenesulfonamide (PB622). A suspension of 1 in 2M aq. HCI/EtOH (1 mL/1 mL) was stirred 2h at 80°C, then 15h at 50°C. The mixture was partitioned between EtOAc and saturated aq. Na2CO3, the aqueous phase was extracted with EtOAc once. The combined organic layers were dried with Na2SO4 and concentrated under reduced pressure to afford the pure amine 23 as a pale yellow solid (20.8 mg, 99%). Rf (EtOAc/cyclohexane, 1/1, v/v) = 0.65. 1H NMR (CD3OD, 200 MHz): <5 1 .00 - 0.77 (m, 3H, H21), 1 .73 - 1 .18 (m, 8H, H17, HIS, H19 and H20), 2.39 (t, J = 6.8 Hz, 2H, Hi 6) , 6.73 (s, 1 H, H2), 7.08 - 6.92 (m, 1 H, Hs), 7.20 (t, J = 8.1 Hz, 1 H, Hs), 7.51 - 7.35 (m, 4H, Haro), 7.76 - 7.62 (m, 2H, Haro). 13C NMR (CD3OD, 50 MHz): <5 14.4, 20.0, 23.6, 29.6, 29.7, 32.5, 90.3, 95.2, 103.5, 120.0, 121 .4, 123.5, 128.2 (2C), 130.2, 130.3, 132.8 (2C), 137.2, 139.1 , 139.6, 150.9, 171 .3. HRMS-ESI (m/z): [M+H]+ calcd for C23H26N3O2S2, 440.1461 ; Found: 440.1472. HPLC (A254): Purity 99.0%; fa: 8.89 min (method 3).
[0195] Combination of a compound of formula (I) and different anticancer treatments
[0196] Material and Methods:
[0197] Cell lines and reagents: The different cell lines were purchased from ATCC. The tumor cell lines were maintained at 37C° and 5% CO2 in humidified atmosphere and grown in DMEM, high glucose, GlutaMAX™ Supplement, pyruvate growth media supplemented with 10% of fetal bovine serum, (ThermoFisher) under normal growing conditions. Cells were treated with the indicated anticancer agent and/or BPR001 -615 at the indicated concentrations and time. All drugs were dissolved in DMSO.
[0198] Proliferation analysis: Cell proliferation was measured using WST-1 reagent from Abeam (#ab65473). At Day 0, cells were plated in 96-well tissue culture plate. At day 1 , the cells were starved in serum (100pL/well). At Day 2 cells were treated with different drugs or DMSO at the indicated concentrations, in quadruplicates. 48h post treatment, WST-1 reagent (10pL/well) was added. The plate was read at TO at 450 nm on a Multiskan FC Counter (ThermoScientific), then every hour until O.D reach 1 .0. Cell proliferation is expressed as percent of the absorbance after subtraction to background (TO).
[0199] Cells were treated for 48 h before the WST-1 assay with variable concentration of BPR001 - 615 and different concentration of the indicated anticancer agent.
[0200] To evaluate the effect of BPR001 -615 alone or in combination with another anticancer agent, we compared observed and expected responses obtained from the combination treatment. Bliss model was used to predict the combined effect of each drug.
[0201] The expected effect (Eexp) of the combination was estimated from each separate drug effect. “SE” corresponds to a significant efficacy of the combination of BPR001 -615 with the identified anticancer agent.
[0202] Antibodies and Reagents Antibody against HMGB1 (#Ab18256), antibody against Annexin A1 (#Ab214486) and antibody against Calreticulin (#Ab2907) were purchased from Abeam (Paris, France). Anti-rabbit IgG and HRP-linked Antibody (#7074S) were purchased from Cell Signaling (Paris, France). Secondary antibody Texas Red goat (#T6391 ) was purchased from ThermoFisher (Paris, France). Oxaliplatin (OXP, Cas 61825-94) and Mitoxanthrone (MTX, Cas 70476-82) were used to induce immunogenic cell death. Fixable viability stain 510 (#564406) was purchased from BD Horizon (Paris, France).
[0203] Detection of HMGB1 Release A375 and CT26 cells were plated at 180 000 cells/ml in 6 wellplates in medium with 2% FBS. The medium was changed after 24h and treatment was added to the cells. Oxaliplatin (OXP) was used at 50pM and 10OpM and BPR001 -615 was used at 3pM, 5pM and 7.5pM. 48h post treatment, supernatants were collected, dying tumor cells were removed by centrifugation and supernatants were isolated and frozen immediately at -20°C. Quantification of HMGB1 and Annexin A1 in the supernatants were assessed by western blot by loading 45pl of the supernatant supplementing with loading buffer. BSA detectable by ponceau staining was used as a loading control.
[0204] Detection of ATP Release A375 cells and U2OS cells were plated at 10 000 cells/ml in 96 well plates with medium supplemented with 2% FBS. 24h later, treatment was added onto the cells (50pL/well) according to the manufacturer’s instructions. Mitoxanthrone (MTX) was used as positive control at 5pM, 10pM and 12.5pM. BPR001 -615 was used in dose response from 2.5pM to 15pM. Following the treatment, RealTime-Glo Extracellular ATP Assay Reagent was added. Mix plate by orbital shaking (300-500rpm) for 30-45 seconds to ensure homogeneity. Quantification of ATP release was assessed by RealTime-Glo Extracellular ATP Assay (Promega, #GA5010).
[0205] Evaluation of calreticulin expression by flow cytometry A375 cells were plated at 180000 cells/ml in 6-well plates in full medium supplemented with 2% FBS. 24h later, indicated compounds were added to the cells. BPR001 -615 was used at indicated concentrations. We used flow cytometry to detect calreticulin as described in Liu et al, Methods in Enzymology, 632 :1 -13, 2020.
[0206] Results:
[0207] BPR001 -615 alone has cytotoxic effects on cancer cell lines associated with ICD properties triggering the 3 hallmarks of ICD in dose-response:
[0208] BPR001 -615 is a BiP(GRP78) Inhibitor inducing ER Stress and CHOP induction (Figure 1 A).
[0209] BPR001 -615 triggers ER Stress-induced & CHOP mediated cancer cells death (Figure 1 B).
[0210] BPR001 -615 triggers calreticulin cell surface exposure on multiple cancer cell lines (Figure 1 C).
[0211] BPR001 -615 triggers HMGB1 and AnnexinAI release (Figure 2A).
[0212] BPR001 -615 triggers extracellular ATP release on multiple cancer cell lines (Figure 2B).
[0213] These data overall show that BPR001 -615 kills tumor cells through ER stress induced apoptosis, autophagy & ICD.
[0214] The combination of a compound of formula (I) such as BPR001 -615 with another anticancer treatment allows obtaining a synergistic anticancer effect:
[0215] The results are summarized in the Tables below:
[0216] These results show that compounds of Formula I such as BPR001 -615 have the ability to remarkably potentiate the effects of anticancer treatments including chemotherapy, targeted therapies and immunotherapies by triggering Immune Cell Death markers for treatments that would trigger no or not enough ICD markers leading to DAMP signal. They have strong anticancer properties and target GRP78. The combination therapy provided a CHOP-mediated cytotoxic effect on cancer cells via endoplasmic-reticulum (ER) Stress induction that is significantly higher than that obtained with each treatment alone.
[0217] Combination treatment regimen of BPR001 -615 together with various chemotherapies 5-FU, Oxaliplatin, Docetaxel or Irinotecan metabolite SN38 were further investigated in the various gastric cancer cell lines at different concentrations using WST-1 cell proliferation assay. The effects of these combination therapies are summarized in Table 1 :
[0218] Table 1 . Summary of in vitro combination synergy observed at BPR001 -615 concentration with various chemotherapies in gastric adenocarcinoma cancers. All combinatory effects in chemotherapy dose response are presented in Figure 3.
[0219] Treatment with BPR001 -615 at 2, 2.5 and 5 pM in combination with chemotherapies, was shown to exert a synergetic effect with chemotherapy in inducing cell death of GC cell lines (human). These results demonstrate an improved therapeutic potential when combining the standard-of-care chemotherapies with BPR001 -615.
[0220] To support this, in vivo combination treatments with targeted and chemotherapy on GC cell lines in mice were undertaken.
[0221 ] Combination of a compound of formula (I) with Trastuzumab in the NCI-N87 model grown in PBMC-humanized NXG mice
[0222] Material and Methods
[0223] A total of 40 female NXG mice aged 5-8 weeks were used for the study (Janvier Laboratories). Mice were allowed 7-days acclimatization before entering the study. Animals were housed in IVC cages (up to 5 per cage) with individual mice identified by tail mark. All animals were allowed free access to a standard certified commercial diet and sanitized water during the study. The holding room was maintained under standard conditions: 20-24° C, 45-65% humidity and a 12h light/dark cycle. NCI-N87 cells (5 x106 1 :1 with matrigel) were implanted on the rear dorsum of female NXG mice. When tumors reached approximately 150 mm3 animals were assigned to treatment 4 groups (7 mice/group): (i) vehicle (15% Kolliphor HS15, 10% PEG400, 5% Ethanol, 70% ultrapure water); (ii) BPR001 -615 (150 mg/kg, Per Os twice a day (BiD) - 12hrs gap); (iii) Trastuzumab 10mg/kg, intravenous (IV), day 1 and day 16; (iv) combination BPR001 -615 150 mg/kg and Trastuzumab 10mg/kg. PBMCs from 2 donors were isolated, processed and dosed IV to the animals immediately prior to therapeutic dosing on the same day as treatment to maximize the treatment window time.
[0224] Tumors were measured three times per week using digital calipers. The length, width and depth of the tumor were measured and used to calculate the tumor volume. The bodyweight of all mice on the study were also measured and recorded three times weekly.
[0225] As efficacy was observed, at termination tumors from this group were excised and digested to a single cell suspension before being stained using the following antibody immune cell panels for FACS analysis.
[0226] Results
[0227] Treatment with BPR001 -615 150mg/kg BID alone or in combination with trastuzumab was well tolerated. BPR001 -615 alone or in combination with trastuzumab decreased intratumoral GRP78
concentrations, with a more significant decrease observed when the drugs are used in combination (see Figure 4).
[0228] Treatment with BPR001 -615 150mg/kg BID alone or in combination with trastuzumab resulted in significant and similar decrease of NCI-N87 tumor growth in PBMC-humanized animals (see Figure 5). Combination animals treated have a tendency to a better decrease in tumor volume that treated animals with trastuzumab to those from animals treated with BPR001 -615 alone.
[0229] FACS analysis of immune cell infiltration into tumor tissue at termination of the study indicated that BPR001 -615 treatment resulted in a range of immune-mediated anti-tumoral effects that are likely to be involved in the efficacy seen and the potentializing of the trastuzumab when used in combination. CD3+ T cells and NK cell infiltration is significantly increased in the case of treatment by combination of BPR001 -615 and trastuzumab versus trastuzumab alone, particularly for NK cell while Treg infiltration is decreased (Figure 6).
[0230] BPR001-615 induces tumor cell death in combination with other treatments
[0231] An overview of the nonclinical in vitro and in vivo pharmacology studies of BPR001 -615 in combination with chemotherapy and immune checkpoint inhibitors in colorectal cancer models is provided in Table 2.
0232] Table 2. Overview of the completed in vitro and in vivo pharmacology studies for BPR001 - 615 (or BPR001 -615 in the table) in combination with chemotherapy and immune checkpoint inhibitors in colorectal cancer model. a-PD1 : Anti-PD1 ; BID: Bis in die; IP: Intraperitoneal; N/A: Not applicable; PO: Per Os; ‘Vehicle for BPR001 -615 is 15% Kolliphor HS15, 10% PEG400, 5% EtOH, 70% water, vehicle for a-PD1 is PBS
[0233] A combination treatment regimen of BPR001 -615 together with chemotherapy drug SN38, Irinotecan metabolite, was investigated in the CT26 cancer cell lines at different drug concentrations using cell proliferation assay. The effects of this combination therapy are presented in Figure 7. Treatment with BPR001 -615 at 2.5 pM, in combination with chemotherapy, was shown to exert a synergetic effect in inducing cell death of CT26 cancer cell lines. These results demonstrate an improved therapeutic potential when combining the standard of care chemotherapy with BPR001 - 615.
[0234] This was further investigated in vivo using immune checkpoint inhibitors. A combination treatment regimen consisting of BPR001 -615 together with an anti-programmed cell death protein 1 antibody (a-PD1 ) was investigated in C57BL/6J female mice subcutaneously implanted with MC38 cancer cells. Four groups (n =10 mice/group) were included in the study to evaluate the effect of BPR001 -615 administered orally at a sub-optimal dose in combination with a-PD1 (intraperitoneal injection; IP), on overall survival. The efficacy of BPR001 -615 in combination with a-PD1 was mainly assessed through survival and tumor size evolution. Results of the in vivo assessment are presented in Figure 8. The overall survival graph shows an improvement in all treatment groups. Notably, the combination of BPR001 -615 with a-PD1 , now standard-of-care for many types of cancers including gastric cancer, yields the most pronounced improvement with one more mouse cured compared to the treatment alone. The results of this study suggest that BPR001 -615 could result in synergistic effects when used alongside existing immunotherapies.
[0235] In vitro study with another compound of formula (I) and a compound not covered by formula
[0236] To confirm the effect of compounds of formula I, combination of BPR001 -615 with cisplatin and irinotecan and compound PB673 (N-(4-(3-((4-(4-phenylbut-1 -yn-1 - yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide) with cisplatin and irinotecan where further analyzed in different cancer cell lines. Both compounds BPR001 -615 and PB673 show equivalent synergistic effects with both chemotherapies (Figure 9).
[0237] The inventors have also compared the effects of BPR001 -615 vs. HA15 in combination with several chemotherapies (cisplatin, docetaxel, irinotecan) in different cancer cell lines. In combination with each chemotherapy, BPR001 -615 shows a superior effect compared to HA15, demonstrating the superiority of compounds of formula I in combination with anticancer agents compared to other compounds (Figure 10).
[0238] BPR001-615’s in vivo efficacy on human NCI-N87 gastric tumours
[0239] The efficacy of BPR001 -615 on gastric tumour growth was evaluated using PBMC-humanized NXG female mice that were subcutaneously implanted with the GC cell line NCI-N87 (1 x 107 in Matrigel 1 :1 ). When tumour volume reached around 150 mm3), PBMCs from two donors were isolated, processed and dosed IV to the animals prior to therapeutic dosing. BPR001 -615 was administered orally twice a day with 12h between administration until day 28 at 150 mg/kg. The study
was also conducted on 7 mice treated with the standard-of-care targeted therapy Trastuzumab (a monoclonal antibody targeting HER2), administered on day 1 and day 16.
[0240] During the dosing period, a good tolerability profile was observed with no body weight loss. The results show a significant inhibition of tumour growth in the mice treated with BPR001 -615 as single agent, reaching up 40% tumour growth inhibition (Figure 1 1 ). The mice treated with BPR001 - 615 showed similar tumour growth inhibition to the one treated with Trastuzumab.
[0241] BPR001 -615 is as efficient as targeted therapy standard-of-care Trastuzumab administered as single agent. This efficacy is demonstrated in BiP (or GRP78) low expressing tumours extending the potential of patients susceptible to respond to the treatment, therefore, to address more than 50% of BiP positive patients, which is far higher than HER2-positive patients population targeted by Trastuzumab (-20%).
[0242] This finding allows envisaging the use of the compounds of the disclosure, in monotherapy as in combination with anticancer agents, for all GRP78 overexpressing tumors, not only for patients high GRP78 tumors but also for low GRP78 tumors independently from HER2 status. [0243] In this context, plasmatic GRP78 can also be used as a biomarker for patients’ selection based on expression of plasmatic GRP78 (low or high).
Claims
[Claim 1] A combination of an anticancer treatment with a compound of formula (I):
[Formula I] :
wherein
Ri represents H, methyl, phenyl or acetyl;
R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4, for use in the treatment of cancer in a patient in need thereof.
[Claim 2] The combination for use according to claim 1 , wherein said anticancer treatment is selected from chemotherapeutic agents, targeted therapies and immunotherapeutic agents.
[Claim 3] The combination for use according to claim 1 or 2, wherein said anticancer treatment is an immunotherapeutic agent selected from immune checkpoint inhibitors.
[Claim 4] The combination for use according claim 3, wherein said immune checkpoint inhibitor is an anti-PD-1 or an anti-PDL1 antibody.
[Claim 5] The combination for use according any one of claims 1 to 4, wherein said compound of formula (I) is selected from the group consisting of
N-(4-(3-((4-(Oct-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-(Hex-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-(3-Hydroxyprop-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-(Cyclohexylethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-((trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-(dec-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((4-(4-phenylbut-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((3-(Oct-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((3-(3-Hydroxyprop-1 -yn-1 -yl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((3-((Trimethylsilyl)ethynyl)phenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(4-(3-((3-Ethynylphenyl)sulfonamido)phenyl)thiazol-2-yl)acetamide;
N-(3-(2-(Methylamino)thiazol-4-yl)phenyl)-4-(oct-1 -yn-1 -yl)benzenesulfonamide;
4-(Oct-1 -yn-1 -yl)-N-(3-(2-(phenylamino)thiazol-4-yl)phenyl)benzenesulfonamide; and
N-(3-(2-Aminothiazol-4-yl)phenyl)-4-(oct-1 -yn-1 -yl)benzenesulfonamide.
[Claim 6] The combination for use according to any one of claims 1 to 5, wherein said compound of formula (I) is BRP001 -615 of formula (II):
[Formula II]:
[Claim 7] The combination for use according to any one of claims 1 to 6, wherein said cancer is selected from the group consisting of breast cancer, bladder cancer, cervical cancer, colorectal cancer, pancreatic cancer, head and neck cancer, Hodgkin lymphoma, liver cancer, lung cancer, kidney cancer, skin cancer, esophagus cancer and stomach cancer.
[Claim 8] The combination for use according to any one of claims 1 to 7, wherein said cancer is colorectal cancer, pancreatic cancer, esophagus cancer or stomach cancer.
[Claim 9] The combination for use according to any one of claims 1 to 7, wherein said cancer is skin cancer, preferably melanoma.
[Claim 10] The combination for use according to any one of claims 1 to 9, wherein said cancer is a non-immunogenic tumor.
[Claim 11] The combination for use according to any one of claims 1 to 10, wherein said cancer is resistant to immunotherapy.
[Claim 12] The combination for use according to claim 11 , wherein said cancer is resistant to immune checkpoint inhibitors.
[Claim 13] The combination for use according to any one of claims 1 to 12, wherein said patient has been identified as overexpressing GRP78.
[Claim 14] The combination for use according to any one of Claims 1 to 12, wherein said patient has a high GRP78 tumor or a low GRP78 tumor.
[Claim 15] A kit of parts comprising:
- an anticancer agent; and
- a compound of formula (I):
[Formula I] :
wherein
Ri represents H, methyl, phenyl or acetyl; R2 represents H, or a C2-C10 alkyl, cycloalkyl or hydroxyalkyl; and wherein the alkynyl chain to which R2 is attached is linked to the benzene ring in position 3 or 4.
[Claim 16] The kit of parts according to claim 13 for use in the treatment of cancer in a patient in need thereof.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP24315195.8 | 2024-04-19 | ||
| EP24315195 | 2024-04-19 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025219595A1 true WO2025219595A1 (en) | 2025-10-23 |
Family
ID=91185302
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2025/060790 Pending WO2025219595A1 (en) | 2024-04-19 | 2025-04-18 | Method for combination treatments using alkynylbenzenesulphonamides for cancer therapy |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025219595A1 (en) |
Citations (27)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5811097A (en) | 1995-07-25 | 1998-09-22 | The Regents Of The University Of California | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
| US5855887A (en) | 1995-07-25 | 1999-01-05 | The Regents Of The University Of California | Blockade of lymphocyte down-regulation associated with CTLA-4 signaling |
| US6051227A (en) | 1995-07-25 | 2000-04-18 | The Regents Of The University Of California, Office Of Technology Transfer | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
| US6207157B1 (en) | 1996-04-23 | 2001-03-27 | The United States Of America As Represented By The Department Of Health And Human Services | Conjugate vaccine for nontypeable Haemophilus influenzae |
| WO2003042402A2 (en) | 2001-11-13 | 2003-05-22 | Dana-Farber Cancer Institute, Inc. | Agents that modulate immune cell activation and methods of use thereof |
| US6682736B1 (en) | 1998-12-23 | 2004-01-27 | Abgenix, Inc. | Human monoclonal antibodies to CTLA-4 |
| US6984720B1 (en) | 1999-08-24 | 2006-01-10 | Medarex, Inc. | Human CTLA-4 antibodies |
| WO2007045996A1 (en) | 2005-10-19 | 2007-04-26 | INSERM (Institut National de la Santé et de la Recherche Médicale) | An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method |
| WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
| US7488802B2 (en) | 2002-12-23 | 2009-02-10 | Wyeth | Antibodies against PD-1 |
| US7605238B2 (en) | 1999-08-24 | 2009-10-20 | Medarex, Inc. | Human CTLA-4 antibodies and their uses |
| WO2010036959A2 (en) | 2008-09-26 | 2010-04-01 | Dana-Farber Cancer Institute | Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor |
| WO2010089411A2 (en) | 2009-02-09 | 2010-08-12 | Universite De La Mediterranee | Pd-1 antibodies and pd-l1 antibodies and uses thereof |
| WO2010117057A1 (en) | 2009-04-10 | 2010-10-14 | 協和発酵キリン株式会社 | Method for treatment of blood tumor using anti-tim-3 antibody |
| US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
| WO2011066342A2 (en) | 2009-11-24 | 2011-06-03 | Amplimmune, Inc. | Simultaneous inhibition of pd-l1/pd-l2 |
| WO2011082400A2 (en) | 2010-01-04 | 2011-07-07 | President And Fellows Of Harvard College | Modulators of immunoinhibitory receptor pd-1, and methods of use thereof |
| US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
| WO2011155607A1 (en) | 2010-06-11 | 2011-12-15 | 協和発酵キリン株式会社 | Anti-tim-3 antibody |
| WO2011159877A2 (en) | 2010-06-18 | 2011-12-22 | The Brigham And Women's Hospital, Inc. | Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions |
| WO2011161699A2 (en) | 2010-06-25 | 2011-12-29 | Aurigene Discovery Technologies Limited | Immunosuppression modulating compounds |
| US8168757B2 (en) | 2008-03-12 | 2012-05-01 | Merck Sharp & Dohme Corp. | PD-1 binding proteins |
| US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
| WO2013006490A2 (en) | 2011-07-01 | 2013-01-10 | Cellerant Therapeutics, Inc. | Antibodies that specifically bind to tim3 |
| WO2014150677A1 (en) | 2013-03-15 | 2014-09-25 | Bristol-Myers Squibb Company | Inhibitors of indoleamine 2,3-dioxygenase (ido) |
| WO2017017004A1 (en) * | 2015-07-24 | 2017-02-02 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Substituted hydrophobic benzene sulfonamide thiazole compounds for use in treating cancer |
| WO2023118373A1 (en) * | 2021-12-21 | 2023-06-29 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Benzene sulfonamide thiazole compounds and their use for the treatment of cancers |
-
2025
- 2025-04-18 WO PCT/EP2025/060790 patent/WO2025219595A1/en active Pending
Patent Citations (27)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5855887A (en) | 1995-07-25 | 1999-01-05 | The Regents Of The University Of California | Blockade of lymphocyte down-regulation associated with CTLA-4 signaling |
| US6051227A (en) | 1995-07-25 | 2000-04-18 | The Regents Of The University Of California, Office Of Technology Transfer | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
| US5811097A (en) | 1995-07-25 | 1998-09-22 | The Regents Of The University Of California | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
| US6207157B1 (en) | 1996-04-23 | 2001-03-27 | The United States Of America As Represented By The Department Of Health And Human Services | Conjugate vaccine for nontypeable Haemophilus influenzae |
| US6682736B1 (en) | 1998-12-23 | 2004-01-27 | Abgenix, Inc. | Human monoclonal antibodies to CTLA-4 |
| US7605238B2 (en) | 1999-08-24 | 2009-10-20 | Medarex, Inc. | Human CTLA-4 antibodies and their uses |
| US6984720B1 (en) | 1999-08-24 | 2006-01-10 | Medarex, Inc. | Human CTLA-4 antibodies |
| WO2003042402A2 (en) | 2001-11-13 | 2003-05-22 | Dana-Farber Cancer Institute, Inc. | Agents that modulate immune cell activation and methods of use thereof |
| US7488802B2 (en) | 2002-12-23 | 2009-02-10 | Wyeth | Antibodies against PD-1 |
| US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
| US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
| WO2007045996A1 (en) | 2005-10-19 | 2007-04-26 | INSERM (Institut National de la Santé et de la Recherche Médicale) | An in vitro method for the prognosis of progression of a cancer and of the outcome in a patient and means for performing said method |
| WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
| US8168757B2 (en) | 2008-03-12 | 2012-05-01 | Merck Sharp & Dohme Corp. | PD-1 binding proteins |
| WO2010036959A2 (en) | 2008-09-26 | 2010-04-01 | Dana-Farber Cancer Institute | Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor |
| US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
| WO2010089411A2 (en) | 2009-02-09 | 2010-08-12 | Universite De La Mediterranee | Pd-1 antibodies and pd-l1 antibodies and uses thereof |
| WO2010117057A1 (en) | 2009-04-10 | 2010-10-14 | 協和発酵キリン株式会社 | Method for treatment of blood tumor using anti-tim-3 antibody |
| WO2011066342A2 (en) | 2009-11-24 | 2011-06-03 | Amplimmune, Inc. | Simultaneous inhibition of pd-l1/pd-l2 |
| WO2011082400A2 (en) | 2010-01-04 | 2011-07-07 | President And Fellows Of Harvard College | Modulators of immunoinhibitory receptor pd-1, and methods of use thereof |
| WO2011155607A1 (en) | 2010-06-11 | 2011-12-15 | 協和発酵キリン株式会社 | Anti-tim-3 antibody |
| WO2011159877A2 (en) | 2010-06-18 | 2011-12-22 | The Brigham And Women's Hospital, Inc. | Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions |
| WO2011161699A2 (en) | 2010-06-25 | 2011-12-29 | Aurigene Discovery Technologies Limited | Immunosuppression modulating compounds |
| WO2013006490A2 (en) | 2011-07-01 | 2013-01-10 | Cellerant Therapeutics, Inc. | Antibodies that specifically bind to tim3 |
| WO2014150677A1 (en) | 2013-03-15 | 2014-09-25 | Bristol-Myers Squibb Company | Inhibitors of indoleamine 2,3-dioxygenase (ido) |
| WO2017017004A1 (en) * | 2015-07-24 | 2017-02-02 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Substituted hydrophobic benzene sulfonamide thiazole compounds for use in treating cancer |
| WO2023118373A1 (en) * | 2021-12-21 | 2023-06-29 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Benzene sulfonamide thiazole compounds and their use for the treatment of cancers |
Non-Patent Citations (29)
| Title |
|---|
| "Agnew Chem Intl. Ed. Engl.", vol. 33, 1994, pages: 183 - 186 |
| BENGSCH ET AL., IMMUNITY, vol. 48, no. 5, 2018, pages 1029 - 1045 |
| BRIGNONE ET AL., J. IMMUNOL., vol. 179, 2007, pages 4202 - 4211 |
| CIFRIC ET AL., ANTIOXIDANTS AND REDOX SIGNALING, 2024 |
| DATABASE MEDLINE [online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; 2022, WU WENBING ET AL: "EGCG Enhances the Chemosensitivity of Colorectal Cancer to Irinotecan through GRP78-MediatedEndoplasmic Reticulum Stress.", XP002812107, Database accession no. NLM36147440 * |
| GAJEWSKI ET AL., TUMOR IMMUNE MICROENVIRONMENT IN CANCER PROGRESSION AND CANCER THERAPY, 2017, pages 19 - 31 |
| HALEBLIAN, J PHARM SCI, vol. 64, no. 8, August 1975 (1975-08-01), pages 1269 - 1288 |
| HUANG ET AL., INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY, vol. 11, no. 11, 2018, pages 5223 |
| LOO ET AL., CLIN. CANCER RES., no. 18, 15 July 2012 (2012-07-15), pages 3834 |
| MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 489 |
| MILLET ANTOINE ET AL: "Discovery and Optimization of N -(4-(3-Aminophenyl)thiazol-2-yl)acetamide as a Novel Scaffold Active against Sensitive and Resistant Cancer Cells", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, no. 18, 22 September 2016 (2016-09-22), US, pages 8276 - 8292, XP055916898, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.6b00547 * |
| NATURE REVIEWS IMMUNOLOGY, vol. 12, April 2012 (2012-04-01) |
| ONCOLOGY LETTERS, vol. 10, no. 4, 2015, pages 2149 - 2155 |
| PARDOLL, NATURE REV CANCER, vol. 12, 2012, pages 252 - 264 |
| PINTO ALINE FERREIRA ET AL: "Thiazole, Isatin and Phthalimide Derivatives Tested in vivo against Cancer Models: A Literature Review of the Last Six Years", vol. 31, no. 20, 10 July 2023 (2023-07-10), NL, pages 2991 - 3032, XP093200264, ISSN: 0929-8673, Retrieved from the Internet <URL:https://www.eurekaselect.com/216226/article> DOI: 10.2174/0929867330666230426154055 * |
| RONCO CYRIL ET AL: "Structure activity relationship and optimization of N -(3-(2-aminothiazol-4-yl)aryl)benzenesulfonamides as anti-cancer compounds against sensitive and resistant cells", vol. 27, no. 10, 1 May 2017 (2017-05-01), Amsterdam NL, pages 2192 - 2196, XP093200257, ISSN: 0960-894X, Retrieved from the Internet <URL:https://pdf.sciencedirectassets.com/271398/1-s2.0-S0960894X17X00097/1-s2.0-S0960894X17303025/main.pdf?X-Amz-Security-Token=IQoJb3JpZ2luX2VjED8aCXVzLWVhc3QtMSJHMEUCIQCpnBBdxySSVeR8jfExPojV2beqF2EEVoUgt91+ghRG6gIgaue1mezbu5Xxswu8FT8qxo9alG7bwwDF5TfbzW2UrbEqsgUIWBAFGgwwNTkwMDM1NDY4NjUiDGOSzTDYJkkroYG7n> DOI: 10.1016/j.bmcl.2017.03.054 * |
| SAKUISHI ET AL., J. EXP. MED., vol. 207, 2010, pages 2187 - 94 |
| SAMANTA ET AL., SCIENTIFIC REPORTS, vol. 10, no. 1, 2020, pages 1 - 12 |
| SHIMIZU ET AL., PATHOLOGY & ONCOLOGY RESEARCH, vol. 23, no. 1, 2017, pages 111 - 116 |
| STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY- VCH |
| THORNTON ET AL., INTERNATIONAL JOURNAL OF CANCER, vol. 133, no. 6, 2013, pages 1408 - 1418 |
| TONG ET AL., PANCREATOLOGY, vol. 21, no. 7, 2021, pages 1378 - 1385 |
| TRUJILLO ET AL., CANCER IMMUNOLOGY RESEARCH, vol. 6, no. 9, 2018, pages 990 - 1000 |
| WANG ET AL., ELIFE, vol. 8, 2019, pages e49020 |
| WHERRY, E. JOHNMAKOTO KURACHI, NATURE REVIEWS IMMUNOLOGY, vol. 15, no. 8, 2015, pages 486 - 499 |
| WU WENBING ET AL: "EGCG Enhances the Chemosensitivity of Colorectal Cancer to Irinotecan through GRP78-MediatedEndoplasmic Reticulum Stress.", JOURNAL OF ONCOLOGY 2022, vol. 2022, 2022, pages 7099589, ISSN: 1687-8450 * |
| XIA ET AL., JOURNAL OF TRANSLATIONAL MEDICINE, vol. 19, no. 1, 2021, pages 1 - 14 |
| ZHANG ET AL., CLINICAL & EXPERIMENTAL METASTASIS, vol. 23, no. 7, 2006, pages 401 - 410 |
| ZHAO ET AL., DIGESTIVE DISEASES AND SCIENCES, vol. 60, no. 9, 2015, pages 2690 - 2699 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Turner et al. | Inhibition of CRM1-dependent nuclear export sensitizes malignant cells to cytotoxic and targeted agents | |
| Corrales et al. | Innate immune signaling and regulation in cancer immunotherapy | |
| US12297210B2 (en) | Naphthyridine compounds and uses thereof | |
| US10335494B2 (en) | Combination of aurora kinase inhibitors and anti-CD30 antibodies | |
| WO2016196935A1 (en) | Compositions comprising a cancer stemness inhibitor and an immunotherapeutic agent for use in treating cancer | |
| JP7433304B2 (en) | Cinnoline compounds and the treatment of HPK1-dependent disorders such as cancer | |
| JP7246309B2 (en) | Oxabicycloheptane for modulating immune responses | |
| WO2023196565A1 (en) | Methods of treating cancer | |
| US20200399377A1 (en) | Combination cancer therapy with anti-cancer agents and antibodies targeting a complex comprising non-classical hla-i and neoantigen | |
| JP2020517629A5 (en) | ||
| KR20180100652A (en) | A combination of a chromene compound and a second activator | |
| WO2021127217A1 (en) | Combination anti-cancer therapies with inducers of iron-dependent cellular disassembly | |
| CN112512520B (en) | P2RX7 modulators in therapy | |
| JP7636322B2 (en) | Madracine derivative compounds, compositions and their uses for treating cancer - Patents.com | |
| WO2025219595A1 (en) | Method for combination treatments using alkynylbenzenesulphonamides for cancer therapy | |
| JP2021523098A (en) | How to regulate antigenicity to enhance recognition by T cells | |
| EP4604939A1 (en) | Combination therapy for the treatment of cancer | |
| JP2020524677A (en) | Combination therapy including targeted therapeutic agents | |
| EP3791185B1 (en) | Selection of patients for combination therapy | |
| US20210361680A1 (en) | Inositol-based immunotherapies | |
| WO2024221005A2 (en) | Methods for inhibiting resistance to immune blockade inhibitor therapy | |
| WO2025114411A1 (en) | New method to treat brain or neurologic disorders | |
| WO2024033400A1 (en) | Sk2 inhibitor for the treatment of pancreatic cancer | |
| HK40050163B (en) | Selection of patients for combination therapy |
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: 25720394 Country of ref document: EP Kind code of ref document: A1 |