WO2025122271A1 - Méthodes de transplantation de cellules souches hématopoïétiques allogénique et traitement du cancer - Google Patents
Méthodes de transplantation de cellules souches hématopoïétiques allogénique et traitement du cancer Download PDFInfo
- Publication number
- WO2025122271A1 WO2025122271A1 PCT/US2024/054062 US2024054062W WO2025122271A1 WO 2025122271 A1 WO2025122271 A1 WO 2025122271A1 US 2024054062 W US2024054062 W US 2024054062W WO 2025122271 A1 WO2025122271 A1 WO 2025122271A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- patient
- cell
- cell product
- days
- 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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/28—Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/11—T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- Allogeneic hematopoietic stem cell transplantation is a potentially curative treatment for hematologic malignancies.
- the donor is a fully human leukocyte antigen (HLA)-matched donor since this decreases risk of graft rejection and graft-versus-host disease (GvHD) (Cytryn et al. (2020), Clin Hematol Int 2(2): 49-58).
- HLA human leukocyte antigen
- GvHD graft-versus-host disease
- a haploidentical donor shares only one HLA haplotype with the recipient and thus has some degree of HLA mismatch (Id.).
- a patient’s parents and children are all haploidentical and the patient’s full sibling has a 50% chance of being haploidentical (Id.).
- haploidentical transplants have expanded accessibility to stem cell transplantation.
- haploidentical HSCT The success of haploidentical HSCT is, however, hindered by multiple complications and the risk of relapse remains high. There is a 51% risk of relapse one year after HSCT and a 60% risk of relapse two years after HSCT.
- the human leukocyte antigen (HLA) disparity between the donor and the recipient patient can lead to graft rejection, graft versus host disease (GvHD), and delayed immune reconstitution with subsequent infectious complications.
- Immunosuppression regimens that are used to prevent GvHD can, in turn, abrogate the graft versus tumor (GvT) effect of the transplanted HSCs thus increasing the risk of disease relapse.
- the survival of patients after HSCT is only about 50% after 1 year and is less than 40% at two years after haploidentical HSCT.
- Gamma-delta (y8) T-cells are an important and unique subset of T lymphocytes as they can recognize a broad range of antigens without antigen priming and are not major histocompatibility complex (MHC) restricted, meaning they can go from donor to recipient without modification or MHC match. They can target and kill cells directly through their cytotoxic activity or indirectly through the activation of other immune cell types.
- y5 T-cell functional responses are induced by several factors including the recognition of stress antigens, which promotes cytokine production and regulates pathogen clearance, inflammation, and tissue homeostasis in response to stress (e.g., a chemotherapeutic agent environment).
- yd T cells are a unique subset of T lymphocytes that can directly kill malignant cells through a variety of mechanisms including the recognition of tumor and/or stress antigens that are not generally expressed on normal healthy tissue including hematopoietic cells.
- yd T cells have been shown to have an anti-leukemic effect in partially mismatched transplant without increasing the risk of GvHD (Lamb et al. (1996), Journal of Hemotherapy 5: 503-9 and Lamb et al. (1999), Cytotherapy 1: 7-19).
- PCT Publication No. WO2017035375A1 described administration of yd T cells after allogeneic HSCT comprising peripheral blood stem cells (PBSCs) and after in vivo T cell depletion.
- PBSCs peripheral blood stem cells
- allogeneic T cells are immunogenic, or in other words, the cells are ‘‘foreign” to the host’s immune system and trigger an immune response or elimination by the innate immune system (Caldwell et al. (2021), Front. Immunol, doi.org/10.3389/fimmu.2020.618427).
- the risk of acute rejection of the allogeneic cellular therapy can be reduced by using lymphodepl eting chemotherapy prior to infusion of the allogeneic immune cells.
- host immune recovery or immune reconstitution after lymphodepletion is considered a “closing window” on the persistence of the allogeneic cell therapy (Borges et al.
- Such patients may receive a reduced regimen or reduced intensity conditioning, which is a less intensive chemotherapy treatment, which can also result in residual leukemic cells, and which increases the likelihood of relapse.
- a method that can deliver a cellular therapy that can target and eliminate any residual leukemic cells could improve patient outcomes by reducing the rate or incidence of relapse.
- a method that increases in vivo persistence and expansion of allogeneic yd T cells in the long term, even after host immune recovery has the potential to reduce the risk of relapse and improve patient outcome.
- Providing yd T cells which can target and kill cancer cells and can go from donor to recipient, would be beneficial in haploidentical HSCT, such as bone marrow transplantation. Such cells would need to persist in the patient for protection to be maintained when the immune system recovers from the lymphodepletion.
- a yd T cell therapy that increases in vivo persistence and expansion of allogeneic yd T cells in the long term, even after host immune recovery, has the potential to reduce the risk of relapse and improve patient outcome.
- the present invention is based, at least partially, on the discovery that a haploidentical bone marrow transplant method that includes reduced intensity conditioning and administration, after bone marrow transplant (BMT), of gamma-delta (y5) T cells that are at least partially -matched as described herein prevents relapse for at least one year and results in in vivo persistence and expansion of allogeneic yd T cells for at least one year after the BMT.
- BMT bone marrow transplant
- y5 T cells gamma-delta
- the invention encompasses a method of increasing the in vivo persistence and expansion of 78 T cells after allogeneic hematopoietic stem cell transplantation (HSCT), the method comprising the steps of: i. administering to a subject on day 0 a hematopoietic stem cell (HSC) graft, wherein the HSCs are bone marrow HSCs obtained from a haploidentical donor, and wherein the graft has not been subjected to T-cell depletion; ii. after day 0 and before day +10, administering to the subject an agent which provides in vivo T cell depletion; optionally, the agent is cyclophosphamide; iii.
- HSC hematopoietic stem cell
- preparing a 78 T cell product by expanding and activating a population of 78 T cells ex vivo, wherein the population of 78 T cells is obtained from a partially-matched donor; and iv. within 9 days after neutrophil engraftment, administering to the subject the 78 T cell product at a dose of about 5xl0 5 to about 5xl0 6 78 T cells/kg; for example, at a dose of about 2.5xl0 6 cells/kg to about 3.5x10 s cells/kg; wherein the method further comprises administering a reduced intensity conditioning regimen prior to day 0.
- the y8 T cells demonstrate persistence and expansion for at least 6 months or at least one year after HSCT; optionally, wherein the number of yd T cells in a peripheral blood sample obtained from the patient at least 180 days after HSCT is greater than about 15,000 cells/ml.
- the 78 T cell product comprises 0 T cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the subject is suffering from leukemia.
- relapse of the leukemia is prevented (or does not occur or recur) for at least about 6 months, 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years after the allogeneic HSCT.
- the patient is suffering from acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML) or myelodysplastic syndrome (MDS).
- AML acute myeloid leukemia
- ALL acute lymphoblastic leukemia
- CML chronic myeloid leukemia
- MDS myelodysplastic syndrome
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 years or older, 60 years or older, 65 years or older, or 70 years or older.
- the partially-matched donor that the population of y8 T cells is obtained from is the same haploidentical donor that the HSCs are obtained from. In yet further aspects, the partially -matched donor that the population of y8 T cells is obtained is a different donor than the haploidentical donor that the HSCs are obtained from.
- the present invention also includes a method of preventing leukemic relapse in a patient in need thereof for at least one year after allogeneic hematopoietic stem cell transplantation, the method comprising the steps of: i. administering to a subject on day 0 a hematopoietic stem cell (HSC) graft, wherein the HSCs are bone marrow HSCs obtained from a haploidentical donor, and wherein the graft has not been subjected to T-cell depletion; ii. after day 0 and before day +10, administering to the subject an agent which provides in vivo T cell depletion; optionally, the agent is cyclophosphamide; iii.
- HSC hematopoietic stem cell
- preparing a 78 T cell product by expanding and activating a population of 78 T cells ex vivo, wherein the population of y8 T cells is obtained from a partially-matched donor; and iv. within 9 days after engraftment, administering to the subject the y8 T cell product at a dose of about 5x10 5 to about 5x10 6 y8 T cells/kg; for example, at a dose of about 2.5xl0 6 cells/kg to about 3.5xl0 6 cells/kg; wherein the method further comprises administering a reduced intensity conditioning regimen prior to day 0.
- the 78 T cells demonstrate persistence and expansion for at least 6 months or at least one year after HSCT; optionally, wherein the number of 76 T cells in a peripheral blood sample obtained from the patient at least 180 days after HSCT is greater than about 15,000 cells/ml.
- the 78 T cell product comprises a
- relapse is prevented (or does not occur or recur) for at least about 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years after the allogeneic HSCT.
- the leukemia is selected from the group consisting of AML, ALL, CML and MDS.
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 year or older, 60 years or older, 65 years or older, or 70 years or older.
- the partially-matched donor that the population of 78 T cells is obtained from is the same haploidentical donor that the HSCs ae obtained from.
- the partially-matched donor that the population of y8 T cells is obtained is a different donor than the haploidentical donor that the HSCs are obtained from.
- HSCT allogeneic hematopoietic stem cell transplantation
- a method for allogeneic hematopoietic stem cell transplantation comprising the steps of: i. administering to a subject on day 0 a hematopoietic stem cell (HSC) graft, wherein the HSCs are bone marrow HSCs obtained from a haploidentical donor, and wherein the graft has not been subjected to T-cell depletion; ii. after day 0 and before day +10, administering to the subject an agent which provides in vivo T cell depletion; optionally, the agent is cyclophosphamide; iii.
- HSC hematopoietic stem cell transplantation
- preparing a y8 T cell product by expanding and activating a population of y8 T cells ex vivo, wherein the population of y8 T cells is obtained from a partially-matched donor; and iv. within 9 days after engraftment, administering to the subject the y8 T cell product at a dose of about 5xlO 5 to about 5xl0 6 y8 T cells/kg; for example, at a dose of about 2.5xl0 6 cells/kg to about 3.5xl0 6 cells/kg; wherein the method further comprises administering a reduced intensity conditioning regimen prior to day 0.
- the 78 T cells demonstrate persistence and expansion for at least 6 months or at least one year after HSCT; optionally, wherein the number of 78 T cells in a peripheral blood sample obtained from the patient at least 180 days after HSCT is greater than about 15,000 cells/ml.
- the 78 T cell product comprises af> T-cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the subject is suffering from leukemia.
- the patient is suffering from leukemia and relapse of leukemia is prevented for at least about 6 months, 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years after the allogeneic HSCT.
- the leukemia is selected from the group consisting of AML, ALL, CML and MDS.
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 year or older, 60 years or older, 65 years or older, or 70 years or older.
- the partially-matched donor that the population of 78 T cells is obtained is the same haploidentical donor that the HSCs are obtained from.
- the partially -matched donor that the population of y8 T cells is obtained is a different donor than the haploidentical donor that the HSCs are obtained from.
- the invention additionally includes a method of treating leukemia in a subject in need thereof, the method comprising allogeneic hematopoietic stem cell transplantation (HSCT) comprising the steps of: i. administering to a subject on day 0 a hematopoietic stem cell (HSC) graft, wherein the HSCs are bone marrow HSCs obtained from a haploidentical donor, and wherein the graft has not been subjected to T-cell depletion; ii. after day 0 and before day +10, administering to the subject an agent which provides in vivo T cell depletion; optionally, the agent is cyclophosphamide; iii.
- HSC hematopoietic stem cell transplantation
- preparing a 76 T cell product by expanding and activating a population of 78 T cells e vivo, wherein the population of 78 T cells is obtained from a partially-matched donor; and iv. within 9 days after engraftment, administering to the subject the 78 T cell product at a dose of about 5xlO 5 to about 5xl0 6 78 T cells/kg; for example, at a dose of about 2.5xl0 6 cells/kg to about 3.5xl0 6 cells/kg; wherein the method further comprises administering a reduced intensity conditioning regimen prior to day 0.
- the 78 T cells demonstrate persistence and expansion for at least 6 months or at least one year after HSCT; optionally, wherein the number of 78 T cells in a peripheral blood sample obtained from the patient at least 180 days after HSCT is greater than about 15,000 cells/ml.
- the 78 T cell product comprises a(3 T-cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- relapse of the leukemia is prevented for at least about 6 months, 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years after the allogeneic HSCT.
- the leukemia is selected from AML, ALL, CML and MDS.
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 year or older, 60 years or older, 65 years or older, or 70 years or older.
- the partially-matched donor that the population of 78 T cells is obtained is the same haploidentical donor that the HSCs are obtained from.
- the partially-matched donor that the population of 78 T cells is obtained is a different donor than the haploidentical donor that the HSCs are obtained from.
- the invention also includes a method of treating cancer in a subject in need thereof, the method comprising; preparing a 78 T cell product by collecting an apheresis sample or a peripheral blood sample from a partially-matched donor and expanding and activating a population of 78 T cells ex vivo therefrom; administering to said patient the 78 T cell product at an effective dose; wherein the method further comprises administering a reduced intensity conditioning regimen prior to day 0.
- the method comprises administering to the subject an agent which provides in vivo T cell depletion, for example, cyclophosphamide.
- the y8 T cells demonstrate persistence and expansion for at least 6 months or at least one year after administration of the y8 T cells; optionally, wherein the number of yd T cells in a peripheral blood sample obtained from the patient at least 180 days after administration of the y8 T cells is greater than about 15,000 cells/ml.
- the y5 T cell product comprises a T-cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the cancer is leukemia.
- the cancer is a solid tumor, including but not limited to glioma and glioblastoma.
- relapse of the cancer is prevented (or does not occur) for at least about 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years.
- the leukemia is selected from AML, ALL, CML and MDS.
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 year or older, 60 years or older, 65 years or older, or 70 years or older.
- the effective dose of the y8 T cell product is a dose between about 5xlO 5 to about 5xl0 6 y5 T cells/kg; for example, at a dose between about 2.5xl0 6 cells/kg to about 3.5xl0 6 cells/kg.
- the invention is a method of increasing in vivo persistence and expansion of allogeneic y8 T cells in a subject in need thereof, wherein the allogeneic y8 T-cells are obtained from a partially-matched donor, the method comprising administering to the subject the y8 T cells at an effective dose, the method further comprising administering a reduced intensity conditioning regimen prior to day 0.
- administering to the subject an agent which provides in vivo T cell depletion for example cyclophosphamide.
- the y8 T cells demonstrate persistence and expansion for at least 6 months or at least one year after HSCT.
- relapse of the cancer is prevented (or does not occur) for at least about 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years.
- the subject is suffering from cancer.
- the cancer is leukemia.
- the cancer is a solid tumor, including but not limited to, glioma and glioblastoma.
- relapse of the cancer, such as leukemia or solid tumor is prevented (or does not occur) for at least about 1 year, 18 months, 2 years, 30 months, 3 years, 42 months, 4 years, 54 months, or 5 years.
- the leukemia is selected from AML, ALL, CML and MDS.
- the subject is suffering from AML, such as high risk or relapsed AML.
- the subject is 50 years or older, 55 year or older, 60 years or older, 65 years or older, or 70 years or older.
- the effective dose of the 78 T cell product is a dose between about 5xlO 5 to about 5xl0 6 78 T cells/kg; for example, at a dose between about 2.5xl0 6 cells/kg to about 3.5xl0 6 cells/kg.
- FIG. 1 A is a schematic summarizing the sequence of treatments used for the clinical study described in the examples.
- the preparatory regimen of fludarabine + cyclophosphamide + total body irradiation (TBI) begins 6 days before the haploidentical HSCT (day -6) and infusion with the EAGD (the 76 T cell product; “INB-100”) was within 7 days after engraftment.
- FIG. IB is a schematic summarizing the methods used for the clinical study described in the examples.
- BM bone marrow
- CD34+ bone marrow graft
- a preparatory regimen comprising fludarabine at 30 mg/m 2 IV from day -6 to day -2, cyclophosphamide (14.5 mg/kg IV) on days -6 and -5, and total body irradiation (TBI) was administered on day -1 (2 Gy).
- TBI total body irradiation
- EAGD expanded and activated 78 T cells
- graft 2 expanded and activated 78 T cells
- EAGD manufacturing took place between day 0 and day +10.
- the EAGD was prepared by obtaining a leukapheresis sample from the haploidentical donor and expanding and activating the 78 T cells on CliniMACS- Prodigy. The EAGD was then cryopreserved until administration to the patient.
- This 78 T cell product was infused into the recipient at either 1 or 3x 1,000,000 cells/kg concentrations depending upon the cohort.
- FIG. 2 is a swimmer’s plot showing the response over time (days) of the 10 treated patients dosed with the y8 T cell product in the clinical study.
- Day 0 on the x-axis is the day of HSCT.
- 100% of the treated patients remain in morphologic complete remission (mCR) > 12 months, and three patients with high-risk disease remaining relapse- free for greater than 3 years with median follow-up 19.2 months and additionally, with no AML patients having relapsed to date (median follow-up 18.7 months).
- FIG. 3A is a bar graph comparing the y8 T cell count (y8 T cells/ml) in peripheral blood samples obtained from three groups of subjects (HaploCy, Cohort 1 and Cohort 2) at 30, 60, 100, 180 and 365 days after haploidentical BMT.
- y8 T cell counts at 30, 60, 100, 180 and 365 days after haploidentical BMT for patients who received haploidentical BMT and post BMT cyclophosphamide without any y8 T cell infusion (HaploCy); 2) y8 T cell counts (at 30, 60, 100, 180 and 365 days after haploidentical BMT) for patients that received a single dose of IxlO 6 cells/kg y8 T cells (Cohort 1), and 3) y8 T cell counts (at 30, 60, 100, 180 and 365 days after haploidentical BMT) for patients that received a single dose of 3x10 6 cells/kg y8 T cells (Cohort 2); Cohort 2 patients received three-times the y8 T cell dose as Cohort 1.
- FIG. 3B shows the same bar graph of FIG. 3A with a dashed line indicating 21xl0 3 y8 T cells/ml which a previous publication (Minculescu et al. (2019), Front. Immunol. 10: 1997) indicated (at 56 days) as providing a long-term survival benefit when some patients naturally reconstituted higher levels of y6 T cells.
- FIG. 4 are plots of cytokine levels (IFNy, TNFa, IL-6, IL-7 and IL-15) for the dosed patients over time as difference median normal serum levels (Observed-median).
- the IL-6 concentration is higher in Cohort 2 but insufficient to initiate clinical manifestations of cytokine release syndrome (CRS). This is also evidenced by decreases of inflammatory cytokines TNF-a, IFN-y, and IL-7 following transplant and infusion. These data are consistent with a favorable clinical safety profile.
- Initial higher concentrations of serum IL-15 are favorable for homeostatic reconstitution, expansion, and durability of the infused yo T cell population consistent with the observed intermediate and longer term efficacy.
- FIGs. 5A-5C are plots showing absolute counts of T cells, B cells and NK cells, respectively, over time (Day -7, Day 30, Day 60, Day 100, Day 180, Day 365) for the dosed patients.
- T cells recover slowly to low normal values from Day 180.
- B cells recover slowly and patients 101-0012’s B cells recovered after viral infection at 59 and remained at normal levels.
- NK cells recover to low normal to normal levels from day 60 with one outlier.
- FIGs. 6A-C are plots showing T-cell subpopulation recovery and shows absolute counts for CD4+ T cells, CD8+ T cells and y8 T cells, respectively, for the dosed patients.
- CD4 and CD8 T cells recover slowly from day 180.
- a significant increase in y8 T cells are observed for Cohort 2 where patients were dosed with 3xl0 6 cells/kg (FIG. 6C).
- This trend continues onw ard up to Day 365 where y8 T cells are increased as compared to that of Cohort 1 where patients were dosed with 1x10 6 cells/kg.
- FIG. 7 compares T cell subsets in Cohort 1 vs. Cohort 2; specifically, the percentages (as percentage of CD3+ cells) of T cell subsets (CD4+, CD8+ and y8 TCR) for Cohort 1 (top) and Cohort 2 (bottom).
- the percentage of y8 T cells is elevated in Cohort 2 as compared to Cohort 1 and above normal levels from day 30 until day 365, demonstrating their expansion during immune reconstitution. At Day 365, percentage of y8 T cells seem to trend back to normal percentages.
- FIGs. 8A and 8B shows the effector memory profile of CD3+, CD8+ cells for dosed patients over time.
- CD8 T cells seem to shift from primarily CD27 Central Memory profile from Day 30 onwards to a more Effector Memory /TEMRA profile up to Day 365.
- CD8 T cells seem to shift from CD197 Central Memory /Effector Memory profile from Day 30 onwards to primarily an Effector Memory profile up to day 365.
- FIG. 9 shows y8 T cell subpopulations (V81 or V82) for dosed patients.
- y8 T cells consist primarily of V82 cells, indicating durability of the initial y8 T V82 product. From Day 180, y8 T populations seem more variable suggesting broader expansion of the initial therapy as the cells persist.
- ratios, concentrations, doses, amounts, and other numerical data may be expressed herein in a range format. It is to be understood that such a range format is used for convenience and brevity, and thus, should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. To illustrate, a concentration range of “about 0.1% to about 5%” should be interpreted to include not only the explicitly recited concentration of about 0.1 wt. % to about 5 wt.
- % but also include individual concentrations (e.g., 1%, 2%, 3%, and 4%) and the sub-ranges (e.g., 0.5%, 1.1%, 2.2%, 3.3%, and 4.4%) within the indicated range.
- the term “about” can include ⁇ 1%, ⁇ 2%, ⁇ 3%, ⁇ 4%, ⁇ 5%, ⁇ 6%, ⁇ 7%, ⁇ 8%, ⁇ 9%, or ⁇ 10%, or more of the numerical value(s) being modified.
- the phrase “about ‘x’ to ‘y’” includes “about ‘x to about ‘y’”. Numbers, ratios, concentrations, amounts, ranges and other numerical data should be construed as modified by the term “about” unless inconsistent with the context.
- administering is meant introducing a compound, biological materials including a cell population, or a combination thereof, or a composition comprising any of the aforementioned compounds, biological materials (e.g., a cell population), or a combination thereof, of the present invention into a human or animal subject.
- One preferred route of administration of the compounds is intravenous.
- Another preferred route is parenteral.
- Parenteral refers to a route of administration that is associated with injection, including intraorbital, infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intrapulmonary.
- Other exemplary routes of administration of the compounds may be intraperitoneal or intrapleural, or via a catheter to the brain.
- any route of administration such as oral, topical, subcutaneous, peritoneal, intra-arterial, inhalation, vaginal, rectal, nasal, introduction into the cerebrospinal fluid, intracranial, or instillation into body compartments can be used.
- Direct injection into a target tissue site such as a solid tumor is also contemplated.
- intracranial administration of the 78 T-cells for the treatment of a glioma or other intracranial tumor can be used.
- subject and “patient” are used interchangeably herein and refer to a human subject and patient.
- a “haploidentical donor” is one who shares, by common inheritance, one HLA haplotype with the recipient. Such donors are also referred to as “half-matched.”
- a “partially matched donor” is a donor that shares one or more MHC alleles with the recipient patient. “Partially matched” in reference to cells, e.g. y8 T cells, means the cells were obtained from a partially -matched donor. In certain aspects, the partially-matched donor has at least two MHC alleles in common with the recipient patient.
- “Haplomatched” in reference to cells or a graft means the cells or graft are obtained from a haploidentical donor.
- “In vivo persistence” in the context of allogeneic cellular therapy, e.g., allogeneic y8 T cell therapy refers to the survival or maintenance of the cells in the patient after administration of the cellular therapy product (e.g. the y6 T cell product) to the recipient patient.
- “In vivo expansion” in the context of allogeneic cellular therapy, e.g., allogeneic y8 T cell therapy refers to the growth or proliferation of the cells in vivo after administration of the cellular therapy product (e.g. the y8 T cell product) to the recipient.
- Increasing the in vivo persistence and expansion of y5 T cells refers to increasing the persistence and/or expansion of the y8 T cells after infusion or administration.
- the persistence of allogeneic cellular therapies is needed for long-term responses and earlier studies demonstrated a lack of long-term persistence and/or expansion of most allogeneic innate immune cell therapies.
- an increased in vivo persistence and expansion can encompass an increased presence of circulating y8 T cells (e.g., as measured in a peripheral blood sample obtained from the patient) 100 days, 180 days, 365 days, 18 months, 2 years or more after HSCT than a control population or than would be expected based on earlier studies or previous models of immune cell persistence in vivo.
- y5 T-cells refers to a subset of T-cells that express a distinct T-cell receptor (TCR) on their surface.
- TCR T-cell receptor
- the majority of T-cells have a TCR composed of two glycoprotein chains called a- and 0-TCR chains (“ «P T cells”).
- P T cells glycoprotein chains
- the TCR is made up of one y-chain and one 3-chain. This group of T-cells is much less common than aP T-cells.
- the y6 T-cell is classified as an innate immune cell but sits at the nexus of the immune system with properties of both adaptive T-cells and innate immune cells.
- y8 T-cells are unique amongst T-cell types in that they do not require antigen processing and MHC presentation of peptide epitopes in order to identify and kill unhealthy cells.
- y5 T-cells are also not MHC-restricted, meaning they can go from a donor to a recipient without inducing graft versus host disease, or GvHD, from MHC mismatch.
- y5 T-cells are believed to have a prominent role in recognition of lipid antigens, cell-surface antigens and to respond to stress-related antigens such as MIC-A and MIC-B and other ligands of the NKG2D receptor. Upon recognition of an unhealthy cell, they can kill directly through perforin and granzyme, similar to an NK cell.
- V51 and V62 y5 T-cells are the two main subpopulations of y3 T-cells as based on their TCR expression.
- V62 y5 T-cells are typically circulating lymphocytes and constitute the majority of peripheral blood y3 T-cells and perform immune surveillance.
- V81 yo T-cells are generally tissue-resident lymphocytes, abundant in mucosal surfaces and epithelia of the digestive, respiratory and urogenital tracts (Caron et al. (2021), Front Immunol. https://doi.org/10.3389/fimmu.2021.666983).
- y8 T cells include Vy9V82 T-cells and Vy9V81 T-cells.
- Human y8 T-cells can also exhibit an antigen-presenting capacity. Similar to dendritic cells (DCs), blood Vy9V62 T-cells are able to respond to signals from microbes and tumors and prime CD4 + and CD8 + T-cells. y6 T-APCs are believed to cross-present antigens directly to CD8 + T-cells. The intracellular protein degradation and endosomal acidification are significantly delayed in y8 T-cells in comparison to monocyte-derived DCs.
- the antigens are transported across IRAP (Insulin-Regulated Amino Peptidase)-positive early and late endosomes, and their processing consists of an export to the cytosol for degradation by the proteasome before being imported into an MHC-I-loading compartment.
- IRAP Insulin-Regulated Amino Peptidase
- Activated y8 T-cells are able to phagocytose tumor antigens and apoptotic or live cancer cells possibly through the scavenger receptor CD36 in a C/EBPa (CCAAT/enhancer-binding protein a)-dependent mechanism and mount a tumor antigen-specific CD8 + T-cell response.
- y8 T-cells can also induce DC maturation through TNF-a production.
- y8 T-cells can process a wide range of antigens for presentation and stimulate other immune cells. Therefore, y6 T-cells’ role in response to infections or cancer may be leveraged to design new strategies in order to improve clinical response of human y8 T-cell-based immunotherapy.
- Increased tumor immunogenicity e.g., increased upregulation of ligands for the NKG2D receptor
- DDR inhibition e.g., PARP inhibition
- a composition, y8 T cell product, cell composition or population of cells can be enriched for the y6 T-cells or the y6 T-cells, for example.
- enriched refers to increasing the total percentage of one or more cytotoxic immune cell types present (e.g., y8 T-cells and/or NK cells) in a sample, relative to the total percentage of the same one or more cell types prior to enrichment, as disclosed herein.
- a sample that is “enriched” for one or more types of cytotoxic immune cell may comprise between about 10% to 100% of the one or more cytotoxic immune cell types in the sample, whereas the total percentage of one or more of the cytotoxic immune cell types in a sample prior to enrichment was, for example, between 0% and 10%.
- an enriched sample compnses at least 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60% ,70%, 80%, 90% or 100%, of one or more types of cytotoxic immune cell.
- Samples may be enriched for one or more cell types using standard techniques, for example, flow cytometry techniques.
- highly enriched refers to increasing the total percentage of one or more cytotoxic immune cell types in a sample such that the one or more cytotoxic immune cell types may comprise between at least about 70% to about 100% of the cytotoxic immune cell type in the sample, whereas the total percentage of that same type of cytotoxic immune cell prior to enrichment was, for example, between 0% and 10%.
- a highly enriched sample comprises at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more of one or more types of cytotoxic immune cell. Samples may be highly enriched for one or more cell types using standard techniques, for example, flow cytometry techniques.
- the “y8 T cell product” is a pharmaceutical composition comprising a pharmaceutically acceptable excipient and expanded and activated y8 T cells, wherein the y8 T cell product is depleted in a.p T cells.
- the yd T cell product or the expanded population of y8 T cells can thus be enriched in y8 T cells and depleted in ap T cells.
- the y5 T cell product can be prepared, for example, by collecting peripheral blood sample or an apheresis product from a partially-matched or haploidentical donor and expanding and activating the population of y8 T cells ex vivo therefrom.
- y8 T cells in “y8 T cell product” are not chimeric antigen receptor (CAR)-y8 T cells.
- the y8 T cell product is prepared by expanding and activating a population of y8 T cells ex vivo.
- the terms “expanded” and “expanding” and the like as used herein mean expansion of one or more cytotoxic immune cells in a sample means to increase in the number of one or more cytotoxic immune cells in a sample by, for example about at least 2- fold, preferably by about 5-fold, preferably by at least 10-fold, preferably about at least 50- fold or more. Expansion of a cytotoxic immune cell population can be accomplished by any number of methods as are known in the art.
- T-cells can be rapidly expanded using non-specific T-cells receptor stimulation (optionally, in the presence of feeder lymphocytes) and either interleukin-2 (IL-2) or interleukin- 15 (IL- 15), with IL-2 and/or zoledronate being preferred.
- the non-specific T-cell receptor stimulus can, for example, include around 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (available from ORTHO-MCNEIL®, Raritan, N.J.).
- T-cells can be rapidly expanded by stimulation of an apheresis sample or peripheral blood mononuclear cells (PBMC) in vitro or ex vivo with one or more antigens (including antigenic portions thereof, such as epitope(s), or a cell) of the cancer, which can be optionally expressed from a vector, such as an human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3 pM MART-1 (26-35,27 L) or gplOO (209-217,210M), in the presence of a T-cell growth factor, such as IL-2 or IL-15 (e.g., at about 300 lU/ml), with IL-2 being preferred.
- HLA-A2 human leukocyte antigen A2
- T-cell growth factor such as IL-2 or IL-15
- the term “depleted in,” “depleting,” or “depletion” means that the number or concentration of a particular cell type in a population of cells has been decreased (for example as the result of a particular manipulation or procedure) from an initial original level to a reduced second level. The term does not require a complete removal of the particular cell type from the population of cells.
- a population of T cells or a y8 T cell product is "depleted in” a[3 T cells if the number or concentration of af> T cells in the composition is decreased as compared to the number or concentration of up T cells originally present, such as in an initial population of cells obtained from a donor.
- post-transplant administration of cyclophosphamide to the patient results in vivo T-cell depletion.
- T-cell depletion when used with respect to an HSC graft refers to a method that removes T-cells from the donor HSC graft (e.g., the bone marrow graft). Such methods are described, for example, in Ho et al. (2001), Blood 98(12): 3192- 3204 and include, for example, negative selection techniques for T-cell depletion such as physical separation and/or antibody-based purging. Physical methods for T-cell depletion include density gradient fractionation, soybean lectin agglutination and E-rosette depletion, and counterflow centrifugal elutriation.
- Antibody-based methods include the use of monoclonal antibodies (e.g., anti-CD6 [T12], anti-CD8 (anti-leu-2), anti-TCRab (T10B9), CAMPATH antibodies, and immunotoxins (e.g., anti-CD5-ricin).
- T-cell depletion methods also include a combination of physical and immunologic methods such as CD34+ selection, e.g., by immunoadsorption column, immunomagnetic beads, and soybean agglutination in combination with a monoclonal antibody.
- HSC graft that “has not been subjected to T- cell depletion” is thus an HSC graft (e.g., the bone marrow graft) that has not been subjected to a T-cell depletion method.
- a graft can also be referred to as “T-cell replete.”
- the graft that has not been subjected to T-cell depletion is a minimally manipulated graft.
- minimally manipulated means that the graft isolated from the haploidentical donor is not subject to methods or procedures that alter its relevant biological characteristics.
- the act of removing a portion of the graft isolated from the donor (such as to produce a yb T cell product) as described herein or other routine steps in the preparation, processing and/or storage of the graft (such as, but not limited to, density-gradient separation, cell selection, centrifugation, and cry opreservation) results in a graft that is still "minimally manipulated" as the sampling does not enrich and/or deplete a particular population of cells.
- the graft isolated from a donor (such as a haploidentical donor) that is minimally manipulated is not subject methods or procedures that enrich and/or deplete a particular population of cells (such as, but not limited to, yb T cells) prior to administration to the subject.
- the yb T-cells can also be derived from human induced pluripotent stem cells (hiPSCs).
- the pluripotent stem cells can, for example, be isolated from the patient having the cancer. In other aspects, the pluripotent stem cells may be isolated from a source other than the patient with cancer.
- the optionally enriched and/or optionally expanded compositions comprising yb T-cells can also comprise natural killer (NK) cells and optionally further comprise other immunocompetent cells including, but not limited to, monocytes, macrophages and dendritic cells.
- NK natural killer
- isolated' and “isolated population” of cells refers to a cell or a plurality of cells removed from the tissue or state in which they are found in a subject.
- the terms may further include cells that have been separated according to such parameters as, but not limited to, cell surface markers, a reporter marker such as a dye or label.
- a myeloablative conditioning regimen consists of a single agent or combination of agents expected to destroy the hematopoietic cells in the bone marrow and results in profound pancytopenia within one to three weeks from the time of administration.
- a nonmyeloablative (NMA) regimen is one that causes minimal cytopenia (but significant lymphopenia) by itself.
- Reduced intensity conditioning (RIC) regimens are an intermediate category of regimens that do not fit the definition of myeloablative or nonmyeloablative. Specific regimens are described, for example, in Negrin et al. (2023), uptodate.com/contents/preparative-regimens-for-hematopoietic-cell-transplantation and Bacigalupo et al. Biol Blood Marro Transplant 15(12): 1628-1633.
- reduced intensity conditioning refers to a conditioning regimen other than a myeloablative conditioning regiment and include reduced-intensity conditioning regimen as well as a nonmyeloablative pre-transplant conditioning regimen.
- Such regimens may utilize less chemotherapy and/or radiation than standard regimens which result in myeloablation (substantial destruction of the patient’s bone marrow to ablate hematopoiesis).
- reduced intensity conditioning can be achieved by using less chemotherapy and/or radiation than the standard myeloablative regimens, for example 50-80% (e.g., 55-75% or 60- 70%) of the amount of a chemotherapeutic commonly used for standard myeloablative conditioning.
- Reduced intensity conditioning regimens can include, for example, administration of the purine analog fludarabine and dose reductions of alkylating agents or total body irradiation. Specific reduced intensity condition regimen are described, for example, in Sengsayadeth et al. (2015), Hematologica 100(7): 859-869.
- a specific reduced intensity condition regimen can, for example, be selected from reduced intensity conditioning with fludarabine and cyclophosphamide; Fludarabine and total body irradiation; fludarabine, cyclophosphamide, and total body irradiation; Fludarabine and busulfan; Fludarabine and melphalan; Fludarabine alone; TBI and cladribine; Fludarabine, cyclophosphamide, and ATG; Fludarabine, araC and ida; Cladribine and araC; and Total lymphoid irradiation and ATG.
- the reduced intensity conditioning regimen comprises fludarabine (e.g., about 30 mg/m 2 IV from day -6 to day -2), cyclophosphamide (e.g., about 14.5 mg/kg IV day -6 and day -5), and total body irradiation (2 Gy on day -1).
- fludarabine e.g., about 30 mg/m 2 IV from day -6 to day -2
- cyclophosphamide e.g., about 14.5 mg/kg IV day -6 and day -5
- total body irradiation 2 Gy on day -1).
- the invention encompasses a method for allogeneic hematopoietic stem cell transplantation (HSCT), a method of treating leukemia, a method of increasing in vivo persistence and expansion of allogeneic y5 T cells administered after allogeneic hematopoietic stem cell transplantation, and a method of preventing leukemic relapse in a patient in need thereof for at least one year after allogeneic hematopoietic stem cell transplantation.
- the methods can comprise the steps of: i.
- HSC hematopoietic stem cell
- the HSCs are bone marrow HSCs obtained from a haploidentical donor, and wherein the graft has not been subjected to T-cell depletion; ii. after day 0 and before day +10, administering to the subject an agent which provides in vivo T cell depletion; optionally the agent is cyclophosphamide; iii. preparing a yS T cell product by expanding and activating a population of yd T cells ex vivo, wherein the population of y8 T cells is obtained from a partially-matched donor; and iv.
- HSC hematopoietic stem cell
- day 0 is the day of the HSCT or bone marrow transplantation.
- day 0 is the day the HSC graft (e.g, the bone marrow graft) is administered to the patient.
- Days after HSCT can be referred to herein with a “+,” for example day +10 is 10 days after HSCT.
- Days before HSCT can be referred to herein with “- ,” for example, “day -5” is 5 days before HSCT.
- the agent that provides in vivo T cell depletion is cyclophosphamide.
- the invention also encompasses a method of treating cancer in a patient in need thereof, the method comprising preparing a y8 T cell product by collecting an apheresis sample from a partially -matched donor, for example, a haploidentical donor, and expanding and activating a population of y8 T cells ex vivo therefrom, wherein the y8 T cell product is enriched for the y5 T cells; administering to said patient the y8 T cell product at an effective dose, the method further comprising administering a reduced intensity conditioning regimen prior to day 0; wherein the y8 T cells display in vivo persistence and expansion such that the number of y5 T cells in a peripheral blood sample obtained from the patient at least 180 days after the administration of the y5 T cell product is greater than 15,000 cells/ml.
- such a method can further comprise administering an agent that provides in vivo T- cell depletion, optionally cyclophosphamide.
- the yb T cell product is administered at a dose of about 5xl0 5 to about 5xl0 6 yd T cells per kilograms (kg) of the weight of the patient (cells/kg).
- the yb T cell product is administered at a dose of about 1x10 6 to about 4x10 6 yb T cells/kg, about 1.5xl0 6 to about 4x10 6 yb T cells/kg, about 2x10 6 to about 4xl0 6 yb T cells/kg, about 2.5xl0 6 yb T cells/kg to about 3.5xl0 6 yb T cells/kg, or about 2,8xl0 6 yb T cells/kg to about 3.2xl0 6 yb T cells/kg.
- the yb T cell product is administered at a dose of about 3xl0 6 yb T cells/kg.
- the yb T-cells are present in the yb T cell product at greater than or equal to about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the total cell population, for example, as determined by flow cytometry. In yet further aspects, the yb T-cells are present in the yb T cell product at greater than or equal to about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the total viable cell population, for example, as determined by flow cytometry.
- the yb T cell product composition comprises yb T-cells and NK cells, wherein the yb T-cells are present at greater than or equal to about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the total cell population or the total viable cell population and the NK cells are present at less than or equal to about 35%, 30%, 25%, or 20% (for example, as determined by flow cytometry).
- the composition comprises the a T-cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the composition comprises the yb T-cells and a.
- the yb T-cells are present at greater than or equal to 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the total cell population or the total viable cell population, for example, as determined by flow cytometry and wherein the composition comprises a T-cells at less than or equal to 5% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the composition comprises the af> T-cells at less than or equal to 4%, 3%, 2% or 1% of the total cell population or the total viable cell population, for example, as determined by flow cytometry.
- the composition comprises the yb T-cells, a T-cells and NK cells, wherein the yb T-cells are present at greater than or equal to 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% of the total cell population or the total viable cell population, for example as determined by flow cytometry, the a.
- T-cells are present at less than or equal to 5% of the total cell population or the total viable cell population
- the NK cells are present at less than or equal to about 35%, 30%, 25%, or 20% of the total cell population or the total viable cell population, as determined by flow cytometry.
- the y8 T cell product comprises the y8 T-cells, a0 T-cells and NK cells, wherein the y6 T-cells are present at greater than or equal to 60%, the a0 T-cells are present at less than or equal to 5%, and the NK cells are present at less than or equal to about 25% of the total cell population or the total viable cell population, as determined by flow cytometry.
- the y8 T cell product comprises the y8 T-cells, a0 T-cells and NK cells, wherein the y8 T-cells are present at greater than or equal to 90%, the aP T-cells are present at less than or equal to 3%, and NK cells are present at less than or equal to 7% of the total cell population or the total viable cell population, as determined by flow cytometry.
- the y8 T cell product comprises the y8 T-cells, a T-cells and NK cells, wherein the y5 T-cells are present at greater than or equal to 95%, the aP T-cells and are present at less than or equal to 2%, and NK cells are present at less than or equal to 3% of the total cell population or the total viable cell population, as determined by flow cytometry.
- the y8 T cell product comprises the y8 T-cells, ap T-cells and NK cells, wherein the y6 T-cells are present at greater than or equal to 99%, and the ap T-cells and the NK cells together are present at less than or equal to 1% of the total cell population or the total viable cell population, as determined by flow cytometry .
- the y8 T cell product can be administered within 21 days after neutrophil engraftment. In certain aspects, the y8 T cell product can be administered within 9 days after neutrophil engraftment. In additional aspects, the y8 T cells are administered within 7 days after neutrophil engraftment. In certain specific aspects, the y8 T cells are administered 3 to 9 days after neutrophil engraftment. In yet additional aspects, the y8 T cell product is administered within 3 to 9 days after neutrophil engraftment. In additional aspects, the y8 T cells are administered within 5 to 9 days after neutrophil engraftment. In certain specific aspects, the y8 T cells are administered within 7 days after neutrophil engraftment.
- the administration of the y8 T cell product within 9 days after neutrophil engraftment is the only administration or infusion of y8 T cells or the y8 T cell product that the patient receives; or in other words, the method comprises only a single administration of the y8 T cell product (or y8 T cells), wherein the single administration is within 9 days (e.g. , within 7 days, or between 3 to 9 days after neutrophil engraftment).
- Neutrophil engraftment is defined as the first of three consecutive days with an absolute neutrophil count greater than or equal to 500/pil. Neutrophil engraftment generally occurs about 15 days to 40 days after bone marrow transplantation.
- the methods described herein increase in vivo persistence and/or expansion of 78 T cells, for example, in vivo persistence and/or expansion of 78 T cells administered after HSCT.
- the increase in in vivo persistence and/or expansion is such that there are at least 10,00076 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 15,000 78 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 16,000 y8 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT. In further aspects, the increase in vivo persistence and/or expansion is such that there are at least 17,000 78 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 18,000 78 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT. In further aspects, the increase in vivo persistence and/or expansion is such that there are at least 19,00078 T cells/ml in a peripheral blood sample obtained from the patient at 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 20,00078 T cells/ml in a peripheral blood sample obtained from the patient 60 days, 100 days, 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 30,000 78 T cells/ml in a peripheral blood sample obtained from the patient 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 40,00078 T cells/ml in a peripheral blood sample obtained from the patient 180 days, 365 days, 18 months, 2 years, or more after HSCT.
- the increase in vivo persistence and/or expansion is such that there are at least 15,00078 T cells/ml in a peripheral blood sample obtained from the patient at least 180 days after HSCT. In further embodiments, the increase in vivo persistence and/or expansion is such that there are at least 20,000 78 T cells/ml in a peripheral blood sample obtained from the patient at least 180 days after HSCT. In yet additional embodiments, the increase in vivo persistence and/or expansion is such that there are at least 30,000 78 T cells/ml in a peripheral blood sample obtained from the patient at least 180 days after HSCT.
- the increase in in vivo persistence and/or expansion is such that the amount of circulating 78 T cells as measured in a peripheral blood sample obtained from the patient (e.g., 78 T cells per ml peripheral blood) is greater the mean amount measured in a control population of patients (e.g., 78 T cells per ml peripheral blood) at the same time point.
- the increase in in vivo persistence and expansion can be such that circulating 78 T cells as measured in a peripheral blood sample obtained from the patient is at least about 10%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100%, 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, or 200%, greater than the mean of the control population.
- the increase in in vivo persistence and expansion is such that circulating 78 T cells as measured in a peripheral blood sample obtained from the patient is at least about 1.5 times, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, 9 times, 10 times, 15 times, 20 times, 25 times, 30 times, 35 times, 40 times, 45 times, or 50 times more than the mean of the control population.
- Appropriate control populations can be determined by the skilled artisan.
- the control population can be a population of patients that received haploidentical bone marrow transplant with post-transplant cyclophosphamide but did not receive administration of 78 T cells or the 78 T cell product.
- the control population is a population treated using the same HSCT method as the patient being treated except that the control population is not administered 78 T cells or the 78 T cell product.
- the patient treated according to a method described herein can be 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient is suffering from or diagnosed with acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML), acute myeloid leukemia (AML), or myelodysplastic syndrome (MDS).
- ALL acute lymphoblastic leukemia
- CML chronic myeloid leukemia
- AML acute myeloid leukemia
- MDS myelodysplastic syndrome
- the patient is suffering from relapsed ALL, relapsed CML or relapsed AML.
- relapsed ALL, relapsed AML, or relapsed CML the patient had reached remission but then had a relapse of the disease.
- the patient is suffering from refractory ALL, refractory AML, or refractory CML.
- the patient does not have a TP53 mutation.
- the patient is suffering from or diagnosed with AML.
- AML patients have been categorized into favorable-, intermediate- and adverse/high-risk groups based on their molecular and cytogenetic profiles (Awada et al. (2022), Cancers (Basel) 14(17): 4166). These classifications are based on the cytogenetic and molecular features at the time of diagnosis (Id.).
- the AML can, for example, be AML with intermediate or high risk features.
- the patient is suffering from high risk AML or AML with high risk features.
- High risk AML is a biologically distinct subset of disease and includes a large percentage of adult AML (Schiller (2013), Hematology Am Soc Hematol Educ Program 2013 (1): 201-208).
- High risk AML is characterized by certain cytogenetic and molecular features, and cluster around patients over age 60, patients with antecedent hematological disorders, patients with a high risk karyotype and/or expression of mutated flt3, kit, or other molecular marker(s) (Id ). Many, if not most, AML patients over 60 are high risk.
- the patient suffering from high risk AML has a high risk cytogenetic abnormality (high risk cytogenetic AML) and/or high risk karyotype.
- high risk cytogenetic abnormalities and/or high risk karyotypes include at least on or more of the following: del 7, trisomy 8, FLT3 internal tandem duplications, secondary or therapy related AML, mixed phenotype AML, del5 and del 17; complex karyotypes (6;9)(p23 ;q34.
- the patient has high risk AML and has an FTL3 mutation such as FLT3 internal tandem duplication.
- the patient has intermediate-risk AML.
- the patient suffering from high risk AML or AML with intermediate or high risk features is 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient is high risk if they are suffering a comorbidity, for example, according to the HCT- comorbidity index (HCT-CI) described, for example, in Artz (2016), Hematology Am Soc Hematol Educ Program 2016 (1): 99-105, and has advanced age (e.g., is 50 years or older, 55 years or older, 60 years or older, or 65 years or older).
- the patient is suffering from or diagnosed with AML and does not have a TP53 mutation.
- the patient being treated according to a method described herein is a patient suffering from or diagnosed with AML that is 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient being treated is a patient suffering from AML that is 50 years or older, 55 years or older, 60 years or older, or 65 years or older, and has a comorbidity, for example according to the HCT-comorbidity index (HCT- CI) described, for example, in Artz (2016), Hematology Am Soc Hematol Educ Program 2016 (1): 99-105.
- HCT-comorbidity index HCT-comorbidity index
- the patient being treated is a patient suffering from AML that is 50 years or older, 55 years or older, 60 years or older, or 65 years or older and has a high risk cytogenetic abnormality (high risk cytogenetic AML) and/or high risk karyotype.
- AML high risk cytogenetic abnormality
- the patient being treated is suffering from or diagnosed with high risk MDS.
- High risk MDS patients are MDS patients with a high risk of evolving to AML and with a poor survival prognosis. Whether a MDS patient is a “high risk” patient can be determined using the Revised International Prognostic Scoring System for MDS, or IPSS- R (Greenberg et al., Blood. 2012 Sep. 20; 120(12): 2454-2465, incorporated herein by reference).
- the IPSS-R takes into account the patient's bone marrow blast percentage, cytogenetic abnormalities, the number and extent of cytopenias in order to place a patient in a prognostic category.
- a patient with an IPSS-R score greater than 4.5 is considered to be a “high-risk” MDS patient.
- the patient suffering from high risk MDS is 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient is suffering from or diagnosed with MDS and does not have a TP53 mutation.
- the patient being treated is suffering from or diagnosed with chronic phase CML.
- the patient suffering from high risk MDS is 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient is suffering from or diagnosed with CML and does not have a TP53 mutation.
- the patient being treated is suffering from or diagnosed with ALL, including, but not limited to, ALL with high-risk features.
- the patient suffering from high risk MDS is 50 years or older, 55 years or older, 60 years or older, or 65 years or older.
- the patient is suffering from or diagnosed with ALL and does not have a TP53 mutation.
- the methods described herein can be used to treat a patient suffering from or diagnosed with a condition selected from one of the following: (i) patients with neoplastic hematological disorders with indication of allogeneic transplant according to the National Comprehensive Cancer Network (NCCN) or other standard guidelines as follows; (a) Acute lymphoblastic leukemia [ALL] with high-risk features or relapsed disease (relapsed ALL); (b) Hodgkin or Non-Hodgkin lymphoma [HL or NHL] : relapsed disease where remission duration is less than 1 year, relapse after previous autologous transplant, or failure to achieve complete response (CR) with chemotherapy; and (c) Myeloid malignancy (such as for example acute myeloid leukemia [AML] with intermediate/high-risk features (per NCCN criteria) or relapsed disease, OR chronic myeloid leukemia [CML] 29 in hematological remission or chronic phase); (li) myeloid disorder (such as for example myel
- the methods can include administration of an agent which provides in vivo T cell depletion or the use of an in vivo T cell depletion protocol after HSCT. Any such protocol known in the art may be used.
- the agent that provides T cell depletion is cyclophosphamide.
- the T cell depletion protocol is cyclophosphamide administered between days +1 and +10.
- the cyclophosphamide is administered at dose between 30 and 70 mg/kg or 50 mg/kg.
- the is cyclophosphamide is administered at a dose of about 30 and 70 mg/kg or 50 mg/kg at days +3 and +4.
- the methods described herein can prevent or reduce the risk of relapse.
- the patient remains in morphologic complete remission (mCR) for at least six months, at least one year, at least 18 months, at least 2 years, at least 30 months, at least 3 years, at least 4 years, at least 5 years, or at least 10 years after the allogeneic HSC transplantation.
- the risk of relapse at one year after HSCT is less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, or less than about 2%.
- the patient survives for at least one year, at least 18 months, at least 2 years, or at least 30 months, at least 3 years, at least 42 months, at least 4 years or more after the allogeneic HSC transplantation.
- the risk of death at one year after HSCT is less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, or less than about 2%.
- the event-free survival rate or probability for subjects treated using the methods described herein at 6 months is greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, or greater than about 95%.
- overall survival rate for example, at 6 months or 1 year, is greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, or greater than about 95%.
- the subject treated with the method described herein exhibits event-free survival, relapse-free survival, or survival to at least 6 months, or at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 years after HSCT.
- a GvHD prophylaxis regimen is used with the allogeneic bone marrow transplantation.
- the GvHD prophylaxis regimen is any known in the art.
- the GvHD prophylaxis regimen provides for a decreased number of agents and/or a reduced concentration of one or more agents to be used, wherein the agent provides for suppression of the immune system.
- the GvHD prophylaxis regimen is one of the following: (i) CELLCEPT® (my cophenolate mofetil) will be given as 15 mg/kg orally (PO) 3 times daily (maximum daily dose of 3 gm) starting day +5 to day +35.
- Tacrolimus can be continued until day +100 and then may be tapered to none by day +180 if there is no evidence of active GvHD; or (iii) Tacrolimus can be given as 0.03 mg/kg/day (dosing may be adjusted as is standard for drug interactions with concurrent medications) IV infusion beginning on day +5 and converted to oral tacrolimus when PO intake is tolerated. Tacrolimus can be continued until day +50 and then may be tapered to none by day +100 if there is no evidence of active GvHD.
- the methods of the present disclosure may be used in conjunction with growth factor treatment.
- any growth factor treatment regimen may be used.
- the growth factor is granulocytecolony stimulating factor (G-CSF).
- G-CSF is administered from day +5 to about day +20 or from day +5 to about day +15.
- the G-CSF is administered at about 5 mcg/kg on day +5 after transplant until neutrophil engraftment.
- the method further comprises a combination of (i) administering a GvHD prophylaxis regimen after day 0; and (ii) administering a growth factor after day 0.
- the methods described herein can be used in the treatment of any condition, cancer or malignancy for which HSCT is used and/or for the treatment of a hematologic malignancy.
- the invention encompasses a method of treating cancer in a patient in need thereof, the method comprising preparing a 76 T cell product by collecting an apheresis sample from a partially -matched donor, for example, a haploidentical donor, and expanding and activating a population of 78 T cells ex vivo therefrom, administering to said patient the 78 T cell product at an effective dose, the method optionally further comprising administering a reduced intensity conditioning regimen prior to day 0.
- Specific cancers and malignancies that can be treated include, for example, hematologic malignancy or cancer.
- Such cancers include hematopoietic cancers (myelodysplastic cancer), myelodysplastic syndromes, pancreatic cancer, head and neck cancer, skin tumors, Minimal Residual Disease (MRD) among: acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), adult B-cell malignancies, including CLL (chronic lymphocytic leukemia), CML (chronic myelogenous leukemia), non-Hodgkin lymphoma (NHL), pediatric B-cell malignancies, including B lineage ALL (acute lymphocyte leukemia), multiple myeloma, lung cancer, breast cancer, ovarian cancer, prostate cancer, colon cancer, melanoma or other hematological and solid tumors, or any combination thereof.
- ALL acute Lymphocytic Leukemia
- AML Acute Myelogenous Leukemia
- NHL chronic lymphocytic leukemia
- CML chronic myelogen
- condition cancer or malignancy is selected from the group consisting of neoplastic hematological disorders with indication of allogenic transplant; acute lymphoblastic leukemia (ALL) with high-risk features; relapsed ALL; Hodgkin lymphoma (HL); Non-Hodgkin lymphoma (NHL); relapsed HL or NHL wherein remission duration is less than one year; relapsed HL or NHL after previous autologous transplant; HL or NHL with failure to achieve complete response (CR) with chemotherapy; myeloid malignancy; acute myeloid leukemia (AML) with intermediate/high-risk features; relapsed AML; chronic myeloid leukemia (CML) in hematological remission or chronic phase; myeloid disorder; myelodysplastic syndrome (MDS) with intermediate/high-risk features; refractory MDS; MDS with myeloproliferative disorder; primary or secondary MDS with high-risk features; refractory MDS; a
- the invention is a method of treating a solid tumor, the method comprising preparing a yd T cell product by collecting an apheresis sample from a partially-matched donor, for example, a haploidentical donor, and expanding and activating a population of y6 T cells ex vivo therefrom; administering to said patient the y8 T cell product at an effective dose, the method further comprising administering a reduced intensity conditioning regimen prior to day 0.
- the solid tumor is a sarcoma, a carcinomas, or a lymphoma.
- the solid tumor is a glioma and includes, for example, glioblastoma.
- the y8 T cells demonstrate persistence and expansion for at least 6 months or at least one year after administration of the y8 T cells; optionally, wherein the number of y8 T cells in a peripheral blood sample obtained from the patient at least 180 days after administration of the y8 T cells is at least about 15,000 cells/ml.
- the y8 T cells demonstrate persistence and expansion such that the number of y5 T cells in a peripheral blood sample obtained from the patient at least 180 days after administration of the y8 T cells is at least about 20,000 cells/ml.
- the y8 T cells demonstrate persistence and expansion such that the number of y8 T cells in a peripheral blood sample obtained from the patient at least 180 days after administration of the y5 T cells is at least about 30,000 cells/ml.
- the patient is suffering from leukemia.
- the invention includes a method for allogeneic hematopoietic stem cell transplantation (HSCT) and a method of increasing in vivo persistence and expansion of allogeneic y8 T cells administered after allogeneic hematopoietic stem cell transplantation wherein the patient suffering from leukemia.
- the invention also encompasses a method for allogeneic hematopoietic stem cell transplantation (HSCT), a method of treating leukemia and a method of preventing leukemic relapse in a patient in need thereof for at least one year after allogeneic hematopoietic stem cell transplantation.
- HSCT allogeneic hematopoietic stem cell transplantation
- the methods of treatment described can be used to treat or reduce a cancer or tumor.
- reducing a cancer “inhibition of cancer,” “inhibiting cancer, “‘preventing cancer recurrence,” and similar terms and are used interchangeably herein and refer to one or more of a reduction in the size or volume of a tumor mass, a decrease in the number of metastasized tumors in a subject, a decrease in the proliferative status (the degree to which the cancer cells are multiplying) of the cancer cells, prevention of recurrences of previous tumors or the development of new metastases, prevention of relapse, prevention of posttransplant relapse, achieving remission, and the like.
- measurable residual disease can be assessed during and/or after the treatment regimen.
- MRD assessment in AML can include multiparameter flow cytometry-based MRD (MFC- MRD) and/or molecular MRD (Mol-MRD) assessed by qPCR (Dohner et al. (2022), Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN, Blood 140(12): 1345-1377; the contents of which are expressly incorporated by reference herein).
- MFC- MRD multiparameter flow cytometry-based MRD
- Mol-MRD molecular MRD assessed by qPCR
- the active agents described herein e.g., HSCs, 76 T cells, chemotherapeutic agents are each administered in a therapeutically effective amount or effective amount.
- therapeutically effective amount or an “effective amount” in the context of the administration of an agent or composition to a subject, refers to an amount capable of having a detectable, positive effect on any symptom, aspect, or characteristic of a disease, disorder or condition, when administered to the subject; the agent or composition can be administered either alone or as part of a pharmaceutical composition and either in a single dose or as part of a series of doses.
- the therapeutically effective amount or effective amount can be ascertained by measuring relevant physiological effects, and it can be adjusted in connection with the dosing regimen and diagnostic analysis of the subject's condition, and the like.
- a therapeutically effective amount or an effective amount can refer to that amount which has the effect of (1) reducing the size of a tumor (i.e. tumor regression), (2) inhibiting (that is, slowing to some extent, preferably stopping) aberrant-cell division, for example cancer cell division, (3) preventing or reducing the metastasis of cancer cells, (4) relieving to some extent (or, preferably, eliminating) one or more symptoms associated with a pathology related to or caused in part by unregulated or aberrant-cellular division, including for example, cancer, (5) increasing the survival or life expectancy of the subject, and/or (6) decreasing the risk of relapse.
- an “effective amount” can also be an amount that results in desirable PD and PK profiles and/or desirable immune cell profiling or reconstitution upon administration of the therapeutically active compositions of the invention.
- An “effective amount” can also be an amount that achieves a recited effect or result; for example, an effective amount of a chemotherapeutic agent that, alone or when in combination with another agent, can be an amount that reduces the size of a tumor and/or increases stress antigen expression on the tumor cells, and/or has a cytotoxic effect.
- treating or “treatment” of a disease (or a condition or a disorder) as used herein refer to inhibiting the disease (slowing or arresting its development), providing relief from the symptoms or side-effects of the disease (including palliative treatment), preventing or delaying recurrence, and causing regression of the disease.
- these terms also mean that the life expectancy of an individual affected with a cancer may be increased or that one or more of the symptoms of the disease will be reduced.
- “treating” also includes enhancing or prolonging an anti-tumor or anti-cancer response in a subject.
- preventing or “prevent” in the context of cancer or leukemic relapse as used herein means to inhibit relapse or ensure that relapse does not occur.
- chemotherapeutic agent refers to a compound or a derivative thereof that can interact with a cancer cell, thereby reducing the proliferative status of the cell and/or killing the cell for example, by impairing cell division or DNA synthesis, or by damaging DNA, effectively targeting fast dividing cells.
- chemotherapeutic agents include, but are not limited to, alkylating agents (e.g., cyclophosphamide, ifosfamide, temozolomide, chlorambucil, bendamustine); metabolic antagonists (e.g., methotrexate (MTX), trimetrexate (TMTX), 5 -fluorouracil or derivatives thereof); a substituted nucleotide; a substituted nucleoside; DNA demethylating agents (also known as antimetabolites; e.g., azacitidine); antitumor antibiotics (e.g., mitomycin, adriamycin); plant-derived antitumor agents (e.g., vincristine, vindesine, TAXOL®, paclitaxel, abraxane); cisplatin; carboplatin; oxaliplatin; etoposide; and the like.
- alkylating agents e.g., cyclophospham
- Such agents may further include, but are not limited to, the anti-cancer agents trimetrexate (TMTX); temozolomide (TMZ); anthracy cline; raltitrexed; S-(4-Nitrobenzyl)-6-thioinosine (NBMPR); 6-benzyguanidine (6-BG); a nitrosourea (rabinopyranosyl-N-methyl-N-nitrosourea (Aranose), Carboplatin, Carmustine (BCNU, BiCNU), Chlorozotocin, Bendamustine, Bleomycin; Cyclophosphamide, Chlorambucil, Cisplatin, Cladribine, Cytarabine, Doxorubicin, Etoposide, Ethylnitrosourea (ENU), Fludarabine, Fotemustine, Gemcitabine, Ifosfamide, Lomustine (CCNU), Methotrexate, Mitoxantrone, Nimustine, N
- Cytarabine is an antineoplastic drug of formula used as a sterile solution for intravenous, intrathecal or subcutaneous administration. Cytarabine injection in combination with other approved anticancer drugs is indicated for remission induction in acute non-lymphocytic leukemia of adults and pediatric patients. It has also been found useful in the treatment of acute non-lymphocytic leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, and the blast phase of chronic myelocytic leukemia. Intrathecal administration of Cytarabine inject on (preservative free preparations only) is indicated in the prophylaxis and treatment of meningeal leukemia.
- Cyclophosphamide (Cytoxan, CY). Cyclophosphamide is a synthetic antineoplastic drug chemically recognized as 2-[bis(2-cholorethyl)amino]tetrahydro-2H-l,3,2- oxazaphosphorine 2-oxide monohydrate.
- Fludarabine Fludarabine phosphate (fludarabine) is an antimetabolite.
- Busulfan (BUSULFEX®) Busulfan is a bifunctional alkylating agent known chemically as dimethanesulfonate ester of 1,4-butanediol.
- Tacrolimus (PROGRAF®). Tacrolimus is a macrolide immunosuppressant produced by Stretocyces Tsukubaensis.
- Mvcophenolate Mofetil (MMF. CELLCEPT®) CELLCEPT® (mycophenolate mofetil) is the 2-morpholinoethyl ester of mycophenolic acid (MPA), an immunosuppressive agent, inosine monophosphate dehydrogenase (IMPDH) inhibitor.
- MPA mycophenolic acid
- IMPDH inosine monophosphate dehydrogenase
- the chemical name for mycophenolate mofetil (MMF) is 2-morpholinoethyl (E)-6-(l,3-dihydro-4-hydroxy-6- methoxy-7- methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate.
- NEUPOGEN® is the trademark name for filgrastim, representing recombinant methionyl human granulocyte colony-stimulating factor (r- methHuG-CSF).
- NEUPOGEN® is a 175 amino acid protein produced by recombinant DNA technology utilizing Escherichia coli (E. coli).
- TBI Total Body Irradiation
- any form of administration of a “combination”, “combined therapy” and/or “combined treatment regimen,” or “co-admimstration” or “co-admmist ering,” or the like refers to administration of at least two therapeutically active drugs or compositions (e.g., administration of the yb T-cells and chemotherapeutic agent, or pharmaceutical compositions thereof), simultaneously or substantially simultaneously in either separate or combined formulations, or sequentially at different times separated by minutes, hours, days, weeks, or months, but in some way act together to provide the desired therapeutic response, for example, as part of the same treatment regimen.
- enhanced refers to an increase in the response or outcome referred to.
- enhancing cytotoxicity refers to increasing cytotoxicity.
- enhanced persistence means increasing persistence.
- the term “enhancing” and the like can also encompass allowing a subject or tumor cell to improve its ability to respond to a treatment disclosed herein.
- An enhanced response can comprise an increase in responsiveness (cytotoxicity and/or persistence) of at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98% or more.
- the enhanced responsiveness can encompass enhanced cytotoxicity to the cancer or tumor and/or enhanced persistence.
- reduced refers to a decrease in the response or outcome referred to.
- reduced cytotoxicity to a normal, non-cancerous cell refers to a decreased cytotoxicity to that normal, non-cancerous cell, for example, as compared with the cytotoxicity to a cancer cell.
- a reduced cytotoxicitycan comprise a decrease in cytotoxicity of at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98% or more.
- cancer as used herein, shall be given its ordinary meaning, as a general term for diseases in which abnormal cells divide without control. Cancer cells can invade nearby tissues and can spread through the bloodstream and lymphatic system to other parts of the body. When normal cells lose their ability to behave as a specified, controlled and coordinated unit, a tumor is formed. Generally, a solid tumor is an abnormal mass of tissue that usually does not contain cysts or liquid areas (some brain tumors do have cysts and central necrotic areas filled with liquid). A single tumor may even have different populations of cells within it, with differing processes that have gone awry. Solid tumors may be benign (not cancerous), or malignant (cancerous). Different types of solid tumors are named for the type of cells that form them.
- Solid tumors are sarcomas, carcinomas, and lymphomas.
- Leukemias (cancers of the blood) generally do not form solid tumors.
- Carcinoma is cancer that begins in the skin or in tissues that line or cover internal organs.
- Glioma is a tumor that arises from the supportive (“gluey“) tissue of the brain, called glia, which helps to keep the neurons in place and functioning well.
- Sarcoma is cancer that begins in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue.
- Leukemia is cancer that starts in blood-forming tissue such as the bone marrow, and causes large numbers of abnormal blood cells to be produced and enter the bloodstream.
- Lymphoma is cancer that begins in the cells of the immune system.
- Representative cancers include, but are not limited to, Acute Lymphoblastic Leukemia (ALL), Adult; Acute Lymphoblastic Leukemia, Childhood; Acute Myeloid Leukemia (AML), Adult; Adrenocortical Carcinoma; Adrenocortical Carcinoma, Childhood; AIDS-Related Lymphoma; AIDS-Related Malignancies; Anal Cancer; Astrocytoma, Childhood Cerebellar; Astrocytoma, Childhood Cerebral; Bile Duct Cancer, Extrahepatic; Bladder Cancer; Bladder Cancer, Childhood; Bone Cancer, Osteosarcoma/Malignant Fibrous Histiocytoma; Glioblastoma, Childhood; Glioblastoma, Childhood; Brain Stem Glioma, Childhood; Brain Tumor, Adult; Brain Tumor, Brain Stem Glioma, Childhood; Brain Tumor, Cerebellar Astrocytoma, Childhood; Brain Tumor, Cerebral Astro
- Hvpopharyngeal Cancer Hypothalamic and Visual Pathway Glioma, Childhood; Intraocular Melanoma; Islet-cell Carcinoma (Endocrine Pancreas); Kaposi's Sarcoma; Kidney Cancer; Laryngeal Cancer; Laryngeal Cancer, Childhood; Leukemia, Acute Lymphoblastic, Adult; Leukemia, Acute Lymphoblastic, Childhood; Leukemia, Acute Myeloid, Adult; Leukemia, Acute Myeloid, Childhood; Leukemia, Chrome Lymphocytic; Leukemia, Chronic Myelogenous; Leukemia, Hairy Cell; Lip and Oral Cavity Cancer; Liver Cancer, Adult (Primary); Liver Cancer, Childhood (Primary ); Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoblastic Leukemia, Adult Acute; Lymphoblastic Leukemia, Childhood Acute; Lymphocytic Leukemia, Chronic; Lymphoma, AIDS
- a tumor can be classified as malignant or benign. In both cases, there is an abnormal aggregation and proliferation of cells. In the case of a malignant tumor, these cells behave more aggressively, acquiring properties of increased invasiveness. Ultimately, the tumor cells may even gain the ability to break away from the microscopic environment in which they originated, spread to another area of the body (with a different environment, not normally conducive to their growth), and continue their rapid growth and division in this new location. This is called metastasis. Once malignant cells have metastasized, achieving a cure or treatment is more difficult. Benign tumors have less of a tendency to invade and are less likely to metastasize.
- compositions described herein can be delivered as a pharmaceutical composition, or made into an implant appropriate for administration in vivo, with appropriate carriers or diluents, which further can be pharmaceutically acceptable.
- appropriate carriers or diluents which further can be pharmaceutically acceptable.
- the means of making such compositions or implants have been described in the art.
- compositions described herein can be formulated into a preparation in semisolid or liquid form, such as a capsule, solution, injection, inhalant, or aerosol, in the usual ways for their respective route of administration.
- Means known in the art can be utilized to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed- release of the composition.
- a pharmaceutically acceptable form is employed which does not ineffectuate the cells.
- the cells as described herein can be made into a pharmaceutical composition containing a balanced salt solution, for example, Hanks' balanced salt solution, or normal saline. Therefore, the invention includes pharmaceutical compositions comprising Y ⁇ T-cells of the present disclosure.
- a pharmaceutical composition described herein can be used alone or in combination with other well-established agents useful for treating cancer, for example, a chemotherapeutic agent as described herein. Whether delivered alone or in combination with other agents, the pharmaceutical composition of the present invention can be delivered via various routes and to various sites in a mammalian, particularly human, body to achieve a particular effect.
- a particular route can provide a more immediate and more effective reaction than another route.
- intradermal delivery may be advantageously used over inhalation for the treatment of melanoma.
- Local or systemic delivery can be accomplished by administration comprising application or instillation of the formulation into body cavities, inhalation or insufflation of an aerosol, or by parenteral introduction, comprising intramuscular, intravenous, intraportal, intrahepatic, peritoneal, subcutaneous, or intradermal administration.
- composition(s) described herein can be provided in unit dosage form wherein each dosage unit, e.g., an injection, contains a predetermined amount of the composition, alone or in appropriate combination with oilier active agents.
- unit dosage form as used herein refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of the composition of the present invention, alone or in combination with other active agents, calculated in an amount sufficient to produce the desired effect, in association with a pharmaceutically acceptable diluent, carrier, or vehicle, where appropriate.
- the specifications for the novel unit dosage forms of the present invention depend on the particular pharmacodynamics associated with the pharmaceutical composition in the particular subject.
- a therapeutically effective amount or sufficient number of the y8 T-cells, administered alone or in combination with an additional therapeutic agent, is introduced into the subject such that a long-term, specific, response is established.
- the response includes inhibition of cancer.
- the response is the reduction in size of a tumor or elimination of tumor growth or regrowth or a reduction in metastasis to a greater degree than would otherwise result in the absence of the treatment with the y8 T-cells or composition thereof.
- the therapeutically effective amount results in at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 100% decrease in tumor size. Accordingly, the therapeutically effective amount takes into account the route of administration and the number of cells should be such that a sufficient number of so as to achieve the desired therapeutic response.
- the amounts of the y8 T-cells of the present disclosure and/or additional cells included in the compositions described herein can vary in different applications.
- the concentration of the cells can be sufficient to provide in the subject being treated at least from about IxlO 5 to about IxlO 10 cells although any suitable amount can be utilized.
- the dosing schedule can be based on well-established cell-based therapies or an alternate continuous infusion strategy can be employed.
- the amounts and dosages described herein provide general guidance to be utilized by the practitioner upon optimizing the method of the present invention for practice of the invention.
- the recitation herein of such ranges by no means precludes the use of a higher or lower amount of a component, as might be warranted in a particular application.
- the actual dose and schedule can vary depending on whether the compositions are administered in combination with other pharmaceutical compositions, or depending on interindividual differences in pharmacokinetics, drug disposition, and metabolism.
- the infused cells are able to kill tumor cells in the recipient.
- the y8 T cells are able to replicate in vivo resulting in long-term persistence that can lead to sustained tumor control.
- the cells administered to the patient, or their progeny persist in the patient for at least four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, twelve months, thirteen months, fourteen months, fifteen months, sixteen months, seventeen months, eighteen months, nineteen months, twenty months, twenty-one months, twenty -two months, twenty- three months, two years, three years, four years, or five years after administration of the cells to the patient.
- the cancer to be treated can be a hematological cancer or a solid tumor.
- the total amount of an agent to be administered in practicing a method of the invention can be administered to a subject as a single dose, either as a bolus or by infusion over a relatively short period of time, or can be administered using a fractionated treatment protocol, in which multiple doses are administered over a prolonged period of time.
- a fractionated treatment protocol in which multiple doses are administered over a prolonged period of time.
- a pharmaceutical composition can be formulated to be compatible with the intended method or route of administration; exemplary routes of administration are set forth herein. Furthermore, the pharmaceutical compositions can be used in combination with other therapeutically active agents or compounds as described herein in order to treat or prevent the diseases, disorders and conditions as contemplated by the present disclosure.
- compositions typically comprise a therapeutically effective amount of one or more agents and one or more pharmaceutically and physiologically acceptable formulation agents.
- suitable pharmaceutically acceptable or physiologically acceptable diluents, carriers or excipients include, but are not limited to, antioxidants (e.g., ascorbic acid and sodium bisulfate), preservatives (e.g., benzyl alcohol, methyl parabens, ethyl or n-propyl, p-hydroxybenzoate), emulsifying agents, suspending agents, dispersing agents, solvents, fillers, bulking agents, detergents, buffers, vehicles, diluents, and/or adjuvants.
- antioxidants e.g., ascorbic acid and sodium bisulfate
- preservatives e.g., benzyl alcohol, methyl parabens, ethyl or n-propyl, p-hydroxybenzoate
- emulsifying agents suspending agents, dispersing agents, solvent
- a suitable vehicle can be physiological saline solution or citrate buffered saline, possibly supplemented with other materials common in pharmaceutical compositions for parenteral administration.
- Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles.
- Typical buffers include, but are not limited to, pharmaceutically acceptable weak acids, weak bases, or mixtures thereof.
- the buffer components can be water soluble materials such as phosphoric acid, tartaric acids, lactic acid, succinic acid, citric acid, acetic acid, ascorbic acid, aspartic acid, glutamic acid, and salts thereof.
- Acceptable buffering agents include, for example, a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2- ethanesulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N- Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), and N-tris[Hydroxymethyl]methyl-3-aminopropanesulfonic acid (TAPS).
- HEPES N-(2-Hydroxyethyl)piperazine-N'-(2- ethanesulfonic acid)
- MES 2-(N-Morpholino)ethanesulfonic acid
- MES 2-(N- Morpholino)ethanesulfonic acid sodium salt
- MOPS 3-(N-Morpholino)propanes
- a pharmaceutical composition After a pharmaceutical composition has been formulated, it can be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations can be stored either in a ready-to-use form, a lyophilized form requiring reconstitution prior to use, a liquid form requiring dilution prior to use, or other acceptable form.
- the pharmaceutical composition is provided in a single-use container (e.g., a single-use vial, ampoule, syringe, or autoinjector (similar to, e.g., an EPIPEN®), whereas a multi-use container (e.g., a multi-use vial) is provided in other embodiments.
- Any drug delivery apparatus can be used to deliver IL-10, including implants (e.g., implantable pumps) and catheter systems, slow injection pumps and devices, all of which are well known to the skilled artisan.
- Depot injections which are generally administered subcutaneously or intramuscularly, can also be utilized to release the polypeptides disclosed herein over a defined period of time. Depot injections are usually either solid- or oil-based and generally comprise at least one of the formulation components set forth herein.
- One of ordinary skill in the art is familiar with possible formulations and uses of depot injections.
- the pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension.
- This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents mentioned herein.
- the sterile injectable preparation can also be a sterile injectable solution or suspension in a non- toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol.
- Acceptable diluents, solvents and dispersion media that can be employed include water, Ringer's solution, isotonic sodium chloride solution, CREMOPHOR ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS), ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures thereof.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium.
- any bland fixed oil can be employed, including synthetic mono- or diglycerides.
- fatty acids such as oleic acid, find use in the preparation of injectables. Prolonged absorption of particular injectable formulations can be achieved by including an agent that delays absorption (e.g., aluminum monostearate or gelatin).
- compositions can be in a form suitable for oral use, for example, as tablets, capsules, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups, solutions, microbeads or elixirs.
- Pharmaceutical compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions, and such compositions can contain one or more agents such as, for example, sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets, capsules and the like contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
- excipients can be, for example, diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, com starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
- diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate
- granulating and disintegrating agents for example, com starch, or alginic acid
- binding agents for example starch, gelatin or acacia
- lubricating agents for example magnesium stearate, stearic acid or talc.
- the tablets, capsules and the like suitable for oral administration can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action.
- a time-delay material such as glyceryl monostearate or glyceryl distearate can be employed. They can also be coated by techniques known in the art to form osmotic therapeutic tablets for controlled release.
- Additional agents include biodegradable or biocompatible particles or a polymeric substance such as polyesters, polyamine acids, hydrogel, polyvinyl pyrrolidone, polyanhydrides, polygly colic acid, ethylene-vinylacetate, methylcellulose, carboxymethylcellulose, protamine sulfate, or lactide/gly colide copolymers, polylactide/glycolide copolymers, or ethylenevinylacetate copolymers in order to control delivery of an administered composition.
- a polymeric substance such as polyesters, polyamine acids, hydrogel, polyvinyl pyrrolidone, polyanhydrides, polygly colic acid, ethylene-vinylacetate, methylcellulose, carboxymethylcellulose, protamine sulfate, or lactide/gly colide copolymers, polylactide/glycolide copolymers, or ethylenevinylacetate copolymers in order to control delivery of an administered composition.
- the oral agent can be entrapped in microcapsules prepared by coacervation techniques or by interfacial polymerization, by the use of hydroxymethylcellulose or gelatin-mi crocapsules or poly (methylmethacrolate) microcapsules, respectively, or in a colloid drug delivery system.
- Colloidal dispersion systems include macromolecule complexes, nano-capsules, microspheres, microbeads, and lipid-based systems, including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Methods for the preparation of the above-mentioned formulations will be apparent to those skilled in the art.
- Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate, kaolin or microcrystalline cellulose, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
- an inert solid diluent for example, calcium carbonate, calcium phosphate, kaolin or microcrystalline cellulose
- water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
- Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture thereof.
- excipients can be suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy-propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents, for example a naturally-occurring phosphatide (e.g., lecithin), or condensation products of an alkylene oxide with fatty acids (e.g., polyoxy-ethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols (e.g., for heptadecaethyleneoxy cetanol), or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol (e.g., polyoxyethylene sorbitol monooleate), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides
- Oily suspensions can be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
- the oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents can be added to provide a palatable oral preparation.
- Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
- a dispersing or wetting agent e.g., kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, ka
- the pharmaceutical compositions can also be in the form of oil-in-water emulsions.
- the oily phase can be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example, liquid paraffin, or mixtures of these.
- Suitable emulsifying agents can be naturally occurring gums, for example, gum acacia or gum tragacanth; naturally occurring phosphatides, for example, soy bean, lecithin, and esters or partial esters derived from fatty acids; hexitol anhydrides, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
- Formulations can also include carriers to protect the composition against rapid degradation or elimination from the body, such as a controlled release formulation, including implants, liposomes, hydrogels, prodrugs and microencapsulated delivery systems.
- a time delay material such as glyceryl monostearate or glyceryl stearate alone, or in combination with a wax, can be employed.
- Suppositories can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
- suitable non-irritating excipient include, but are not limited to, cocoa butter and polyethylene glycols.
- compositions suitable for use in accordance with the invention may be in any format (e.g., sprays for nasal or inhalation use) currently known or developed in the future.
- kits comprising the pharmaceutical compositions typically comprise a therapeutically effective amount of one or more agents used in the combination therapies of the invention described herein.
- Kits typically include a label indicated the intended use of the contents of the kits and instructions for use.
- compositions or a combination of the compositions described herein can be comprised in a kit.
- a chimeric receptor expression construct In a non-limiting example, a chimeric receptor expression construct, one or more reagents to generate a chimeric receptor expression construct, cells for transfection of the expression construct, and/or one or more instruments to obtain autologous cells for transfection of the expression construct (such an instrument may be a syringe, pipette, forceps, and/or any such medically approved apparatus).
- the kits may comprise one or more suitably aliquoted compositions of the present invention or reagents to generate compositions of the invention.
- the components of the kits may be packaged either in aqueous media or in lyophilized form.
- the container means of the kits may include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there are more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
- the kits of the present invention also will typically include a means for containing the chimeric receptor construct and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained, for example.
- kits are generally in the form of a physical structure housing various components, as described below, and can be utilized, for example, in practicing the methods described above.
- a kit can include a composition comprising one or more of the therapeutic agents used in the combination therapy of the invention (e.g. a y8 T cell product) provided in, e.g., one or more sterile containers, which can be in the form of a pharmaceutical composition suitable for administration to a subject.
- the pharmaceutical composition can be provided in a form that is ready for use or in a form requiring, for example, reconstitution or dilution prior to administration.
- the kit can also include buffers, pharmaceutically acceptable excipients, and the like, packaged with or separately the therapeutic agent.
- the kit can contain the several agents separately or they can already be combined in the kit.
- a kit of the invention can be designed for conditions necessary to properly maintain the components housed therein (e.g., refrigeration or freezing).
- a kit can contain a label or packaging insert including identifying information for the components therein and instructions for their use (e.g., dosing parameters, clinical pharmacology of the active ingredient(s), including mechanism(s) of action, pharmacokinetics and pharmacodynamics, adverse effects, contraindications, etc.).
- Each component of the kit can be enclosed within an individual container, and all of the various containers can be within a single package.
- Labels or inserts can include manufacturer information such as lot numbers and expiration dates.
- the label or packaging insert can be, e.g., integrated into the physical structure housing the components, contained separately within the physical structure, or affixed to a component of the kit (e.g., an ampule, syringe or vial).
- Labels or inserts can additionally include, or be incorporated into, a computer readable medium, such as a disk (e.g., hard disk, card, memory disk), optical disk such as CD- or DVD-ROM/RAM, DVD, MP3, magnetic tape, or an electrical storage media such as RAM and ROM or hybrids of these such as magnetic/optical storage media, FLASH media or memory-type cards.
- a computer readable medium such as a disk (e.g., hard disk, card, memory disk), optical disk such as CD- or DVD-ROM/RAM, DVD, MP3, magnetic tape, or an electrical storage media such as RAM and ROM or hybrids of these such as magnetic/optical storage media, FLASH media or memory-type cards.
- the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e g., via an internet site, are provided.
- Example 1 INB 100 - A pilot study of donor-derived, ex vivo expanded/ activated gamma delta T cell (EAGD) infusion following haploidentical hematopoietic stem cell transplantation and post-transplant cyclophosphamide
- EAGD gamma delta T cell
- graft vs. host disease can become more of a significant, even fatal, factor with partial matches.
- T-cells have been shown to be the key player in the post-transplant immune phenomena.
- the majority of T-cells are composed of ap T-cells with a small minority of 78 T-cells, which are known to have the unique ability to kill malignant cells without antigen recognition.
- Gamma-delta (yd) T cells engage heme malignancies via innate sensing of multiple ligands such as PVR, B7-H6, DNAM-1, and the NKG2DL family. This study expands and activates gamma delta T-cells from the peripheral blood to provide innate antitumor effect with minimal risk of GvHD. Safety and impact and/or the rate of GvHD is evaluated.
- Gamma-delta (76) T cells are MHC unrestricted lymphocytes that recognize and lyse malignant cells in allogeneic settings.
- haploidentical transplant with posttransplant cyclophosphamide (PTCy) has reduced the risk of graft-versus-host disease (GvHD), the incidence of relapse remains up to 50% at one year after HSCT.
- Early posttransplant infusion of haploidentical expanded and activated y8T cells (EAGD) may decrease relapse risk through a graft-versus-leukemia (GvL) effect without severe GvHD.
- R2D recommended phase 2 dose
- N 3 (up to 6) patients, single dose of 1 x 10 6 cells/kg
- N 3 (up to 6) patients, single dose of 3 x 10 6 cells/kg (RP2D)
- N 3 (up to 6) patients, single dose of 1 x 10 7 cells/kg
- a bone marrow graft is obtained from a haploidentical donor and administered to the patient on day 0.
- the patient is treated with the preparatory regimen of fludarabine (FLU)/cyclophosphamide (CY)/total body irradiation (TBI).
- FLU fludarabine
- CY cyclophosphamide
- TBI total body irradiation
- EAGD infusion is administered up to 21 days after engraftment, e.g., preferably within seven days after engraftment; neutrophil engraftment is about 15-40 days following HSCT.
- MDS Myelodysplastic syndromes
- ALL Acute lymphocy tic leukemia
- EAGD-related toxi cities were primarily Gl-2, included maculopapular rash (50% incidence with 10% G3 events), acute GvHD (G2 in 70%) and 31% chronic GVHD. Treatment related serious AE’s in 23% of patients included G3 nausea
- FIG. 3A shows in vivo persistence and expansion of the y8 T cells for one year.
- the y8 T cell dose as Cohort 1 was observed at 60 days.
- the circulating y8 T cells (cells/ml peripheral blood) for Cohort 2 are 49-times greater than that of HaploCy patients and 8-times greater than Cohort 1 patients.
- FIG. 3B shows the graph of FIG. 3A with a dashed line indicating 21x10 3 78 T cells/ml which a previous publication (Minculescu et al. (2019), Front. Immunol. 10: 1997) indicated (at 56 days) as providing a long-term survival benefit when some patients naturally reconstituted higher levels of 78 T cells.
- FIG. 4 are plots of cytokine levels (IFNy, TNFa, IL-6, IL-7 and IL-5) for the dosed patients over time as difference median normal serum levels (Observed-median).
- the IL-6 concentration is higher in Cohort 2 but insufficient to initiate clinical manifestations of cytokine release syndrome (CRS). This is also evidenced by decreases of inflammatory cytokines TNF-a, IFN-y, and IL-7 following transplant and infusion. These data are consistent with a favorable clinical safety profile.
- Initial higher concentrations of serum IL-15 are favorable for homeostatic reconstitution, expansion, and durability of the infused 78 T cell population consistent with the observed intermediate and longer term efficacy.
- IL-6 and IL-7 increase post-BMT from Day 26 (pre-infusion) to 30, but drop at day 60, 30 to 34 days after 78 T cell infusion back to normal levels, indicating 78 T cells modulate GvHD.
- IL- 15 increased post-BMT from Day 26 (pre-infusion) to 30, but drop at day 60, 30 to 34 days after 78 T cell infusion back to normal levels, indicating 78 T cells potentially modulate graft- versus-leukemia (GvL).
- T cells recover slowly to low normal values from Day 180.
- B cells recover slowly and patient 101-0012’s B cells recovered after viral infection at 59 and remained at normal levels (FIG. 5B).
- NK cells recover to low normal to normal levels from day 60 with one outlier (FIG. 5C).
- FIGs. 5A-5C show that immune reconstitution followed expected patterns with the T cells recovering to low normal values by Day 180 and B cells and NK cells recover to low normal to normal values by Day 60. This further illustrates that the increase in 78 T cells observed over time is expansion and persistence of the administered therapy, while other immune cell populations are recovering at expected levels.
- CD4 and CD8 T cells recover slowly from day 180.
- the percentage of y8 T cells is elevated in Cohort 2 as compared to Cohort 1 and above normal levels from day 30 until day 365, demonstrating their expansion during immune reconstitution.
- percentage of y8 T cells seem to trend back to normal percentages.
- CD8 T cells seem to shift from primarily CD27 Central Memory profile from Day 30 onwards to a more Effector Memory /TEMRA profile up to Day 365.
- CD8 T cells seem to shift from CD197 Central Memory /Effector Memory profile from Day 30 onwards to primarily an Effector Memory profile up to day 365.
- y8 T cells consist primarily of V82 cells, indicating durability of the initial y8 T V82 product. From Day 180, y8 T populations seem more variable suggesting broader expansion of the initial therapy as the cells persist.
- EAGD cells demonstrated manageable safety with no severe acute GvHD.
- RFS relapse-free survival
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Cell Biology (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Biotechnology (AREA)
- Chemical & Material Sciences (AREA)
- Veterinary Medicine (AREA)
- Hematology (AREA)
- Developmental Biology & Embryology (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Virology (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Organic Chemistry (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
L'invention concerne une méthode de transplantation de cellules souches hématopoïétiques (HSCT) allogénique, une méthode de traitement de la leucémie, une méthode de prévention de la rechute leucémique chez un patient en ayant besoin pendant au moins une année après une transplantation de cellules souches hématopoïétiques allogénique comprenant l'administration d'un produit à base de lymphocytes T γδ après la HSCT allogénique. L'invention concerne également un procédé d'augmentation in vivo de la persistance et de l'expansion de lymphocytes T γδ allogéniques administrés après une transplantation de cellules souches hématopoïétiques allogénique.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363607740P | 2023-12-08 | 2023-12-08 | |
| US63/607,740 | 2023-12-08 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2025122271A1 true WO2025122271A1 (fr) | 2025-06-12 |
| WO2025122271A9 WO2025122271A9 (fr) | 2025-08-28 |
Family
ID=95979595
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/054062 Pending WO2025122271A1 (fr) | 2023-12-08 | 2024-11-01 | Méthodes de transplantation de cellules souches hématopoïétiques allogénique et traitement du cancer |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025122271A1 (fr) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20190183930A1 (en) * | 2015-08-25 | 2019-06-20 | The Uab Research Foundation | Methods for stem cell transplantation |
| WO2023174398A1 (fr) * | 2022-03-17 | 2023-09-21 | Rootpath , Inc. | Lymphocytes synthétiques infiltrant les tumeurs (tils) |
| WO2023205705A2 (fr) * | 2022-04-20 | 2023-10-26 | Bluesphere Bio, Inc. | Lymphocytes t allogéniques pour le traitement de malignités hématologiques |
-
2024
- 2024-11-01 WO PCT/US2024/054062 patent/WO2025122271A1/fr active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20190183930A1 (en) * | 2015-08-25 | 2019-06-20 | The Uab Research Foundation | Methods for stem cell transplantation |
| WO2023174398A1 (fr) * | 2022-03-17 | 2023-09-21 | Rootpath , Inc. | Lymphocytes synthétiques infiltrant les tumeurs (tils) |
| WO2023205705A2 (fr) * | 2022-04-20 | 2023-10-26 | Bluesphere Bio, Inc. | Lymphocytes t allogéniques pour le traitement de malignités hématologiques |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2025122271A9 (fr) | 2025-08-28 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Ayash et al. | Double dose-intensive chemotherapy with autologous marrow and peripheral-blood progenitor-cell support for metastatic breast cancer: a feasibility study. | |
| US6544787B1 (en) | Non-myeloablative/lymphoablative conditioning regimen to induce patient anti-donor unresponsiveness in stem cell transplantation | |
| US11779555B2 (en) | Combination of immunotherapy with local chemotherapy for the treatment of malignancies | |
| KR20170085955A (ko) | 림프종의 치료 | |
| Bruserud et al. | Extracorporeal photopheresis (photochemotherapy) in the treatment of acute and chronic graft versus host disease: immunological mechanisms and the results from clinical studies | |
| CN109641002B (zh) | 血液癌症的组合治疗 | |
| JP7105188B2 (ja) | 幹細胞移植方法 | |
| Persaud et al. | Antibody-drug conjugates plus Janus kinase inhibitors enable MHC-mismatched allogeneic hematopoietic stem cell transplantation | |
| Copelan et al. | Mobilization of peripheral-blood progenitor cells with high-dose etoposide and granulocyte colony-stimulating factor in patients with breast cancer, non-Hodgkin's lymphoma, and Hodgkin's disease. | |
| WO2015154065A1 (fr) | Inhibition d'histone désacétylase 6 pour améliorer la fonction de lymphocytes t pendant une réponse anti-tumorale et une vaccination par peptides tumoraux | |
| WO2018183930A1 (fr) | Procédés d'isolement, d'expansion et d'administration de lymphocytes t cd8+ spécifiques du cancer | |
| JP6802150B2 (ja) | がん治療のための免疫療法 | |
| Okita et al. | Targeting of CD4+ CD25high cells while preserving CD4+ CD25low cells with low-dose chimeric anti-CD25 antibody in adoptive immunotherapy of cancer | |
| Hołowiecki et al. | G-CSF administered in time-sequenced setting during remission induction and consolidation therapy of adult acute lymphoblastic leukemia has beneficial influence on early recovery and possibly improves long-term outcome: a randomized multicenter study | |
| WO2025122271A1 (fr) | Méthodes de transplantation de cellules souches hématopoïétiques allogénique et traitement du cancer | |
| US20080160050A1 (en) | Dendritic cell tumor injection therapy and related vaccine | |
| JP2010508364A (ja) | 樹状細胞腫瘍注射治療及び関連するワクチン | |
| deMagalhaes-Silverman et al. | High-dose chemotherapy and autologous stem cell support followed by post-transplant doxorubicin and taxol as initial therapy for metastatic breast cancer: hematopoietic tolerance and efficacy | |
| Giménez et al. | Intrathecal liposomal cytarabine (DepoCyte®) as treatment for CNS infiltration in an adult woman with acute myeloid leukaemia | |
| Ueda et al. | Mobilization of peripheral blood stem cells (PBSCs) after etoposide, adriamycin and cisplatin therapy, and a multimodal cell therapy approach with PBSCs in advanced gastric cancer | |
| BRPI0712595A2 (pt) | uso de timosina alfa 1 para o tratamento de doenças imunológicas. | |
| CN108368485A (zh) | 具有抗趋除特性的修饰的天然杀伤细胞及其用途 | |
| MXPA99004444A (en) | Conditioning for allogeneic stem cell transplantation | |
| Ball et al. | Targeting of once daily intravenous busulfan as part of conditioning therapy | |
| CN118236361A (zh) | 牛磺酸联合化疗药物在制备抗肿瘤药物中的应用 |
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: 24901295 Country of ref document: EP Kind code of ref document: A1 |