Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Semin Immunol ; 49: 101437, 2020 06.
Article in English | MEDLINE | ID: mdl-33262066

ABSTRACT

Cellular therapies have shown increasing promise as a cancer treatment. Encouraging results against hematologic malignancies are paving the way to move into solid tumors. In this review, we will focus on T-cell therapies starting from tumor infiltrating lymphocytes (TILs) to optimized T-cell receptor-modified (TCR) cells and chimeric antigen receptor-modified T cells (CAR-Ts). We will discuss the positive preclinical and clinical findings of these approaches, along with some of the persisting barriers that need to be overcome to improve outcomes.


Subject(s)
Immunomodulation , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
Int J Cancer ; 146(1): 236-247, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31479522

ABSTRACT

Cetuximab and panitumumab bind the human epidermal growth factor receptor (EGFR). Although the chimeric cetuximab (IgG1) triggers antibody-dependent-cellular-cytotoxicity (ADCC) of EGFR positive target cells, panitumumab (a human IgG2) does not. The inability of panitumumab to trigger ADCC reflects the poor binding affinity of human IgG2 Fc for the FcγRIII (CD16) on natural killer (NK) cells. However, both human IgG1 and IgG2 bind the FcγRII (CD32A) to a similar extent. Our study compares the ability of T cells, engineered with a novel low-affinity CD32A131R -chimeric receptor (CR), and those engineered with the low-affinity CD16158F -CR T cells, in eliminating EGFR positive epithelial cancer cells (ECCs) in combination with cetuximab or panitumumab. After T-cell transduction, the percentage of CD32A131R -CR T cells was 74 ± 10%, whereas the percentage of CD16158F -CR T cells was 46 ± 15%. Only CD32A131R -CR T cells bound panitumumab. CD32A131R -CR T cells combined with the mAb 8.26 (anti-CD32) and CD16158F -CR T cells combined with the mAb 3g8 (anti-CD16) eliminated colorectal carcinoma (CRC), HCT116FcγR+ cells, in a reverse ADCC assay in vitro. Crosslinking of CD32A131R -CR on T cells by cetuximab or panitumumab and CD16158F -CR T cells by cetuximab induced elimination of triple negative breast cancer (TNBC) MDA-MB-468 cells, and the secretion of interferon gamma and tumor necrosis factor alpha. Neither cetuximab nor panitumumab induced Fcγ-CR T antitumor activity against Kirsten rat sarcoma (KRAS)-mutated HCT116, nonsmall-cell-lung-cancer, A549 and TNBC, MDA-MB-231 cells. The ADCC of Fcγ-CR T cells was associated with the overexpression of EGFR on ECCs. In conclusion, CD32A131R -CR T cells are efficiently redirected by cetuximab or panitumumab against breast cancer cells overexpressing EGFR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cetuximab/administration & dosage , Neoplasms/drug therapy , Panitumumab/administration & dosage , Receptors, Antigen, T-Cell/metabolism , Receptors, IgG/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Neoplasms/metabolism , T-Lymphocytes/metabolism
3.
Semin Immunol ; 28(1): 3-9, 2016 02.
Article in English | MEDLINE | ID: mdl-26797495

ABSTRACT

While the clinical progress of chimeric antigen receptor T cell (CAR-T) immunotherapy has garnered attention to the field, our understanding of the biology of these chimeric molecules is still emerging. Our aim within this review is to bring to light the mechanistic understanding of these multi-modular receptors and how these individual components confer particular properties to CAR-Ts. In addition, we will discuss extrinsic factors that can be manipulated to influence CAR-T performance such as choice of cellular population, culturing conditions and additional modifications that enhance their activity particularly in solid tumors. Finally, we will also consider the emerging toxicity associated with CAR-Ts. By breaking apart the CAR and examining the role of each piece, we can build a better functioning cellular vehicle for optimized treatment of cancer patients.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology , Cell Culture Techniques , Genetic Therapy , Humans , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Cell Antigen Receptor Specificity , T-Lymphocytes/transplantation
4.
BMC Cancer ; 19(1): 203, 2019 Mar 06.
Article in English | MEDLINE | ID: mdl-30841880

ABSTRACT

Chimeric antigen receptor T (CAR-T) cells are a promising new treatment for patients with relapsed or refractory hematologic malignancies, including lymphoma. Given the success of CAR-T cells directed against CD19, new targets are being developed and tested, since not all lymphomas express CD19. CD30 is promising target as it is universally expressed in virtually all classical Hodgkin lymphomas, anaplastic large cell lymphomas, and in a proportion of other lymphoma types, including cutaneous T cell lymphomas and diffuse large B cell lymphomas. Preclinical studies with CD30-directed CAR-T cells support the feasibility of this approach. Recently, two clinical trials of CD30-directed CAR-T cells in relapsed/refractory CD30+ lymphomas, including Hodgkin lymphoma, have been reported with minimal toxicities noted and preliminary efficacy seen in a proportion of patients. However, improving the persistence and expansion of CAR-T cells is key to further enhancing the efficacy of this treatment approach. Future directions include optimizing the lymphodepletion regimen, enhancing migration to the tumor site, and combination with other immune regulators. Several ongoing and upcoming clinical trials of CD30-directed CAR-T cells are expected to further enhance this approach to treat patients with relapsed and refractory CD30+ lymphomas.


Subject(s)
Immunotherapy, Adoptive , Ki-1 Antigen/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, CD19 , Antigens, Neoplasm/immunology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Hodgkin Disease/immunology , Hodgkin Disease/therapy , Humans , Lymphoma, Large-Cell, Anaplastic/immunology , Lymphoma, Large-Cell, Anaplastic/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Treatment Outcome
5.
Mol Ther ; 26(12): 2727-2737, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30309819

ABSTRACT

Second-generation (2G) chimeric antigen receptors (CARs) targeting CD19 are highly active against B cell malignancies, but it is unknown whether any of the costimulatory domains incorporated in the CAR have superior activity to others. Because CD28 and 4-1BB signaling activate different pathways, combining them in a single third-generation (3G) CAR may overcome the limitations of each individual costimulatory domain. We designed a clinical trial in which two autologous CD19-specific CAR-transduced T cell products (CD19.CARTs), 2G (with CD28 only) and 3G (CD28 and 4-1BB), were infused simultaneously in 16 patients with relapsed or refractory non-Hodgkin's lymphoma. 3G CD19.CARTs had superior expansion and longer persistence than 2G CD19.CARTs. This difference was most striking in the five patients with low disease burden and few circulating normal B cells, in whom 2G CD19.CARTs had limited expansion and persistence and correspondingly reduced area under the curve. Of the 11 patients with measurable disease, three achieved complete responses and three had partial responses. Cytokine release syndrome occurred in six patients but was mild, and no patient required anti-IL-6 therapy. Hence, 3G CD19.CARTs combining 4-1BB with CD28 produce superior CART expansion and may be of particular value when treating low disease burden in patients whose normal B cells are depleted by prior therapy.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Aged , Combined Modality Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphoma, Non-Hodgkin/diagnosis , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/metabolism , Transplantation, Autologous , Treatment Outcome
6.
Blood ; 127(26): 3350-9, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27207792

ABSTRACT

Hematologic malignancies provide a suitable testing environment for cell-based immunotherapies, which were pioneered by the development of allogeneic hematopoietic stem cell transplant. All types of cell-based therapies, from donor lymphocyte infusion to dendritic cell vaccines, and adoptive transfer of tumor-specific cytotoxic T cells and natural killer cells, have been clinically translated for hematologic malignancies. The recent success of chimeric antigen receptor-modified T lymphocytes in B-cell malignancies has stimulated the development of this approach toward other hematologic tumors. Similarly, the remarkable activity of checkpoint inhibitors as single agents has created enthusiasm for potential combinations with other cell-based immune therapies. However, tumor cells continuously develop various strategies to evade their immune-mediated elimination. Meanwhile, the recruitment of immunosuppressive cells and the release of inhibitory factors contribute to the development of a tumor microenvironment that hampers the initiation of effective immune responses or blocks the functions of immune effector cells. Understanding how tumor cells escape from immune attack and favor immunosuppression is essential for the improvement of immune cell-based therapies and the development of rational combination approaches.


Subject(s)
Adoptive Transfer/methods , Cell- and Tissue-Based Therapy/methods , Hematologic Neoplasms , Receptors, Antigen, T-Cell , Tumor Escape , Animals , B-Lymphocytes/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
8.
Curr Oncol Rep ; 20(9): 73, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30069644

ABSTRACT

PURPOSE OF REVIEW: This review will discuss the challenges facing adoptive cell techniques in the treatment of solid tumors and examine the therapies that are in development for specifically pediatric solid tumors. RECENT FINDINGS: Targeting solid tumors with adoptive cell therapy has been limited by the inhibitory tumor microenvironment and heterogeneous expression of targetable antigens. Many creative strategies to overcome these limitations are being developed but still need to be tested clinically. Early phase clinical trials in neuroblastoma with GD2 CAR T cells are promising but results need to be validated on a larger scale. Most research in other pediatric solid tumors is still in early stages. Adoptive cell therapy represents a useful tool to improve the outcomes of many pediatric solid tumors but significant study is still required. Several clinical trials are ongoing to test therapies that have shown promise in the lab.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Child , Humans
9.
Mol Ther ; 25(3): 580-592, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28187946

ABSTRACT

Immunotherapy with T cells expressing the chimeric antigen receptor (CAR) specific for the CD19 antigen (CD19.CAR-Ts) is a very effective treatment in B cell lymphoid malignancies. However, B cell aplasia and cytokine release syndrome (CRS) secondary to the infusion of CD19.CAR-Ts remain significant drawbacks. The inclusion of safety switches into the vector encoding the CAR is seen as the safest method to terminate the effects of CD19.CAR-Ts in case of severe toxicities or after achieving long-term sustained remissions. By contrast, the complete elimination of CD19.CAR-Ts when CRS occurs may jeopardize clinical responses as CRS and antitumor activity seem to concur. We have demonstrated, in a humanized mouse model, that the inducible caspase-9 (iC9) safety switch can eliminate CD19.CAR-Ts in a dose-dependent manner, allowing either a selective containment of CD19.CAR-T expansion in case of CRS or complete deletion on demand granting normal B cell reconstitution.


Subject(s)
Antigens, CD19/immunology , Caspase 9/metabolism , Cytotoxicity, Immunologic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cell Survival , Gene Expression , Gene Order , Genetic Vectors/genetics , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Mice , Molecular Imaging , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Protein Binding/immunology , Receptors, Antigen, T-Cell/genetics
10.
Mol Ther ; 25(9): 2214-2224, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28602436

ABSTRACT

Targeting disialoganglioside (GD2) on neuroblastoma (NB) with T cells expressing a first-generation chimeric antigen receptor (CAR) was safe, but the cells had poor expansion and long-term persistence. We developed a third-generation GD2-CAR (GD2-CAR3) and hypothesized that GD2-CAR3 T cells (CARTs) would be safe and effective. This phase 1 study enrolled relapsed or refractory NB patients in three cohorts. Cohort 1 received CART alone, cohort 2 received CARTs plus cyclophosphamide and fludarabine (Cy/Flu), and cohort 3 was treated with CARTs, Cy/Flu, and a programmed death-1 (PD-1) inhibitor. Eleven patients were treated with CARTs. The infusions were safe, and no dose-limiting toxicities occurred. CARTs were detectable in cohort 1, but the lymphodepletion induced by Cy/Flu increased circulating levels of the homeostatic cytokine interleukin (IL)-15 (p = 0.003) and increased CART expansion by up to 3 logs (p = 0.03). PD-1 inhibition did not further enhance expansion or persistence. Antitumor responses at 6 weeks were modest. We observed a striking expansion of CD45/CD33/CD11b/CD163+ myeloid cells (change from baseline, p = 0.0126) in all patients, which may have contributed to the modest early antitumor responses; the effect of these cells merits further study. Thus, CARTs are safe, and Cy/Flu can further increase their expansion.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy, Adoptive , Neuroblastoma/immunology , Neuroblastoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adolescent , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Child , Child, Preschool , Cohort Studies , Combined Modality Therapy , Cytokines/blood , Female , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Lymphocyte Count , Lymphocyte Depletion , Male , Molecular Targeted Therapy , Myeloid Cells/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Receptors, Antigen, T-Cell/genetics , Transplantation Conditioning , Treatment Outcome , Young Adult
11.
Immunol Rev ; 257(1): 107-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24329793

ABSTRACT

Investigators developed chimeric antigen receptors (CARs) for expression on T cells more than 25 years ago. When the CAR is derived from an antibody, the resultant cell should combine the desirable targeting features of an antibody (e.g. lack of requirement for major histocompatibility complex recognition, ability to recognize non-protein antigens) with the persistence, trafficking, and effector functions of a T cell. This article describes how the past two decades have seen a crescendo of research which has now begun to translate these potential benefits into effective treatments for patients with cancer. We describe the basic design of CARs, describe how antigenic targets are selected, and the initial clinical experience with CAR-T cells. Our review then describes our own and other investigators' work aimed at improving the function of CARs and reviews the clinical studies in hematological and solid malignancies that are beginning to exploit these approaches. Finally, we show the value of adding additional engineering features to CAR-T cells, irrespective of their target, to render them better suited to function in the tumor environment, and discuss how the safety of these heavily modified cells may be maintained.


Subject(s)
Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens/genetics , Antigens/immunology , Antigens/metabolism , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Transfer Techniques , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
Blood ; 125(25): 3905-16, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25940712

ABSTRACT

Although T cells expressing CD19-specific chimeric antigen receptors (CARs) are a promising new therapy for B-cell malignancies, objective responses are observed at lower frequencies in patients with lymphoma than in those with acute B-cell leukemia. We postulated that the tumor microenvironment suppresses CAR-expressing T cells (CARTs) through the activity of indoleamine 2,3-dioxygenase (IDO), an intracellular enzyme that converts tryptophan into metabolites that inhibit T -: cell activity. To investigate the effects of tumor IDO on CD19-CART therapy, we used a xenograft lymphoma model expressing IDO as a transgene. CD19-CARTs inhibited IDO-negative tumor growth but had no effect on IDO-positive tumors. An IDO inhibitor (1-methyl-tryptophan) restored IDO-positive tumor control. Moreover, tryptophan metabolites inhibited interleukin (IL)-2-, IL-7-, and IL-15-dependent expansion of CARTs; diminished their proliferation, cytotoxicity, and cytokine secretion in vitro in response to CD19 recognition; and increased their apoptosis. Inhibition of CD19-CARTs was not mitigated by the incorporation of costimulatory domains, such as 4-1BB, into the CD19-CAR. Finally, we found that fludarabine and cyclophosphamide, frequently used before CART administration, downregulated IDO expression in lymphoma cells and improved the antitumor activity of CD19-CART in vivo. Because tumor IDO inhibits CD19-CARTs, antagonizing this enzyme may benefit CD19-CART therapy.


Subject(s)
Antigens, CD19/immunology , Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphoma/enzymology , Lymphoma/immunology , T-Lymphocytes/immunology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclophosphamide/pharmacology , Disease Models, Animal , Down-Regulation , Flow Cytometry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/drug effects , Mice , Mice, SCID , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins , T-Lymphocytes/drug effects , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Xenograft Model Antitumor Assays
13.
Blood ; 125(26): 4103-13, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25977584

ABSTRACT

To test the feasibility of a single T-cell manipulation to eliminate alloreactivity while sparing antiviral and antitumor T cells, we infused 12 haploidentical hematopoietic stem cell transplant patients with increasing numbers of alloreplete haploidentical T cells expressing the inducible caspase 9 suicide gene (iC9-T cells). We determined whether the iC9-T cells produced immune reconstitution and if any resultant graft-versus-host disease (GVHD) could be controlled by administration of a chemical inducer of dimerization (CID; AP1903/Rimiducid). All patients receiving >10(4) alloreplete iC9-T lymphocytes per kilogram achieved rapid reconstitution of immune responses toward 5 major pathogenic viruses and concomitant control of active infections. Four patients received a single AP1903 dose. CID infusion eliminated 85% to 95% of circulating CD3(+)CD19(+) T cells within 30 minutes, with no recurrence of GVHD within 90 days. In one patient, symptoms and signs of GVHD-associated cytokine release syndrome (CRS-hyperpyrexia, high levels of proinflammatory cytokines, and rash) resolved within 2 hours of AP1903 infusion. One patient with varicella zoster virus meningitis and acute GVHD had iC9-T cells present in the cerebrospinal fluid, which were reduced by >90% after CID. Notably, virus-specific T cells recovered even after AP1903 administration and continued to protect against infection. Hence, alloreplete iC9-T cells can reconstitute immunity posttransplant and administration of CID can eliminate them from both peripheral blood and the central nervous system (CNS), leading to rapid resolution of GVHD and CRS. The approach may therefore be useful for the rapid and effective treatment of toxicities associated with infusion of engineered T lymphocytes. This trial was registered at www.clinicaltrials.gov as #NCT01494103.


Subject(s)
Caspase 9/genetics , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes/transplantation , Adolescent , Child , Child, Preschool , Female , Flow Cytometry , Genes, Transgenic, Suicide , Haplotypes , Hematopoietic Stem Cell Transplantation/methods , Humans , Lymphoproliferative Disorders/surgery , Male , Middle Aged , Organic Chemicals/therapeutic use , Real-Time Polymerase Chain Reaction , Young Adult
14.
Blood ; 123(24): 3750-9, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24782509

ABSTRACT

Adoptive transfer of T lymphocytes expressing a CD19-specific chimeric antigen receptor (CAR.CD19) induces complete tumor regression in patients with lymphoid malignancies. Although in vivo persistence of CAR-T cells correlates with clinical responses, it remains unknown whether specific cell subsets within the CAR-T-cell product correlate with their subsequent in vivo expansion and persistence. We analyzed 14 patients with B-cell malignancies infused with autologous CAR.CD19-redirected T cells expanded ex vivo using IL-2, and found that their in vivo expansion only correlated with the frequency within the infused product of a CD8(+)CD45RA(+)CCR7(+) subset, whose phenotype is closest to "T-memory stem cells." Preclinical models showed that increasing the frequency of CD8(+)CD45RA(+)CCR7(+) CAR-T cells in the infused line by culturing the cells with IL-7 and IL-15 produced greater antitumor activity of CAR-T cells mediated by increased resistance to cell death, following repetitive encounters with the antigen, while preserving their migration to secondary lymphoid organs. This trial was registered at www.clinicaltrials.gov as #NCT00586391 and #NCT00709033.


Subject(s)
Adult Stem Cells/physiology , Antigens, CD19/genetics , Immunologic Memory , Interleukin-15/pharmacology , Interleukin-7/pharmacology , Lymphoma/therapy , T-Lymphocytes/physiology , Adoptive Transfer/methods , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Animals , Antigens, CD19/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Genetic Therapy/methods , Humans , Lymphocyte Activation/drug effects , Lymphoma/genetics , Lymphoma/immunology , Mice , Mice, SCID , Mice, Transgenic , Receptors, Antigen/genetics , Receptors, Antigen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
15.
Blood ; 123(25): 3895-905, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24753538

ABSTRACT

Adoptive transfer of donor-derived T lymphocytes expressing a safety switch may promote immune reconstitution in patients undergoing haploidentical hematopoietic stem cell transplant (haplo-HSCT) without the risk for uncontrolled graft versus host disease (GvHD). Thus, patients who develop GvHD after infusion of allodepleted donor-derived T cells expressing an inducible human caspase 9 (iC9) had their disease effectively controlled by a single administration of a small-molecule drug (AP1903) that dimerizes and activates the iC9 transgene. We now report the long-term follow-up of 10 patients infused with such safety switch-modified T cells. We find long-term persistence of iC9-modified (iC9-T) T cells in vivo in the absence of emerging oligoclonality and a robust immunologic benefit, mediated initially by the infused cells themselves and subsequently by an apparently accelerated reconstitution of endogenous naive T lymphocytes. As a consequence, these patients have immediate and sustained protection from major pathogens, including cytomegalovirus, adenovirus, BK virus, and Epstein-Barr virus in the absence of acute or chronic GvHD, supporting the beneficial effects of this approach to immune reconstitution after haplo-HSCT. This study was registered at www.clinicaltrials.gov as #NCT00710892.


Subject(s)
Caspase 9/genetics , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/transplantation , Transgenes/genetics , Adolescent , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillosis/prevention & control , Aspergillus fumigatus/immunology , Caspase 9/biosynthesis , Child , Child, Preschool , Enzyme Induction/drug effects , Female , Follow-Up Studies , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Humans , Immunotherapy, Adoptive/methods , Lymphoma, Large-Cell, Anaplastic/immunology , Lymphoma, Large-Cell, Anaplastic/therapy , Male , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/therapy , Organic Chemicals/administration & dosage , Organic Chemicals/therapeutic use , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transplantation, Homologous , Treatment Outcome , Virus Diseases/immunology , Virus Diseases/prevention & control , Virus Diseases/virology
17.
Blood ; 122(20): 3461-72, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24016461

ABSTRACT

Genetically targeted T cells promise to solve the feasibility and efficacy hurdles of adoptive T-cell therapy for cancer. Selecting a target expressed in multiple-tumor types and that is required for tumor growth would widen disease indications and prevent immune escape caused by the emergence of antigen-loss variants. The adhesive receptor CD44 is broadly expressed in hematologic and epithelial tumors, where it contributes to the cancer stem/initiating phenotype. In this study, silencing of its isoform variant 6 (CD44v6) prevented engraftment of human acute myeloid leukemia (AML) and multiple myeloma (MM) cells in immunocompromised mice. Accordingly, T cells targeted to CD44v6 by means of a chimeric antigen receptor containing a CD28 signaling domain mediated potent antitumor effects against primary AML and MM while sparing normal hematopoietic stem cells and CD44v6-expressing keratinocytes. Importantly, in vitro activation with CD3/CD28 beads and interleukin (IL)-7/IL-15 was required for antitumor efficacy in vivo. Finally, coexpressing a suicide gene enabled fast and efficient pharmacologic ablation of CD44v6-targeted T cells and complete rescue from hyperacute xenogeneic graft-versus-host disease modeling early and generalized toxicity. These results warrant the clinical investigation of suicidal CD44v6-targeted T cells in AML and MM.


Subject(s)
Antigens, Neoplasm/immunology , Hyaluronan Receptors/immunology , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/therapy , Molecular Targeted Therapy , Multiple Myeloma/therapy , T-Lymphocyte Subsets/immunology , Animals , Antigens, Neoplasm/genetics , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Line, Tumor/immunology , Cell Line, Tumor/transplantation , Cytotoxicity, Immunologic , Genes, Transgenic, Suicide , Graft vs Host Disease/therapy , Humans , Hyaluronan Receptors/genetics , Interleukin-15/immunology , Interleukin-15/pharmacology , Interleukin-7/immunology , Interleukin-7/pharmacology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/therapy , Lymphocyte Activation , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasm Transplantation , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/immunology , T-Cell Antigen Receptor Specificity , Xenograft Model Antitumor Assays
18.
Blood ; 122(17): 2965-73, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24030379

ABSTRACT

Autologous T cells expressing a CD19-specific chimeric antigen receptor (CD19.CAR) are active against B-cell malignancies, but it is unknown whether allogeneic CD19.CAR T cells are safe or effective. After allogeneic hematopoietic stem cell transplantation (HSCT), infused donor-derived virus-specific T cells (VSTs) expand in vivo, persist long term, and display antiviral activity without inducing graft-vs-host disease; therefore, we determined whether donor VSTs, engineered to express CD19.CAR, retained the characteristics of nonmanipulated allogeneic VSTs while gaining antitumor activity. We treated 8 patients with allogeneic (donor-derived) CD19.CAR-VSTs 3 months to 13 years after HSCT. There were no infusion-related toxicities. VSTs persisted for a median of 8 weeks in blood and up to 9 weeks at disease sites. Objective antitumor activity was evident in 2 of 6 patients with relapsed disease during the period of CD19.CAR-VST persistence, whereas 2 patients who received cells while in remission remain disease free. In 2 of 3 patients with viral reactivation, donor CD19.CAR-VSTs expanded concomitantly with VSTs. Hence CD19.CAR-VSTs display antitumor activity and, because their number may be increased in the presence of viral stimuli, earlier treatment post-HSCT (when lymphodepletion is greater and the incidence of viral infection is higher) or planned vaccination with viral antigens may enhance disease control.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/transplantation , Adenoviridae/immunology , Adult , Antigens, CD19/genetics , Antineoplastic Agents/therapeutic use , B-Lymphocytes/pathology , Child , Cytomegalovirus/immunology , Female , Gene Expression , Herpesvirus 4, Human/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/virology , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Protein Engineering , Recurrence , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transplantation, Homologous
19.
Br J Haematol ; 166(5): 711-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24889268

ABSTRACT

Multiple myeloma (MM) stem cells, proposed to be responsible for the tumourigenesis, drug resistance and recurrence of this disease, are enriched in the cancer stem cell-like side population (SP). Cancer testis antigens (CTA) are attractive targets for immunotherapy because they are widely expressed in cancers but only in limited types of normal tissues. We designed a high throughput assay, which allowed simultaneous relative quantifying expression of 90 CTA genes associated with MM. In the three MM cell lines tested, six CTA genes were over-expressed in two and LUZP4 and ODF1 were universally up-regulated in all three cell lines. Subsequent study of primary bone marrow (BM) from eight MM patients and four healthy donors revealed that 19 CTA genes were up-regulated in SP of MM compared with mature plasma cells. In contrast, only two CTA genes showed a moderate increase in SP cells of healthy BM. Furthermore, knockdown using small interfering RNA (siRNA) revealed that LUZP4 expression is required for colony-forming ability and drug resistance in MM cells. Our findings indicate that multiple CTA have unique expression profiles in MM SP, suggesting that CTA may serve as targets for immunotherapy that it specific for MM stem cells and which may lead to the long-term cure of MM.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplastic Stem Cells/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Apoptosis/immunology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , DNA-Binding Proteins/analysis , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Heat-Shock Proteins/analysis , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , High-Throughput Screening Assays/methods , Humans , Multiple Myeloma/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods
20.
N Engl J Med ; 365(18): 1673-83, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-22047558

ABSTRACT

BACKGROUND: Cellular therapies could play a role in cancer treatment and regenerative medicine if it were possible to quickly eliminate the infused cells in case of adverse events. We devised an inducible T-cell safety switch that is based on the fusion of human caspase 9 to a modified human FK-binding protein, allowing conditional dimerization. When exposed to a synthetic dimerizing drug, the inducible caspase 9 (iCasp9) becomes activated and leads to the rapid death of cells expressing this construct. METHODS: We tested the activity of our safety switch by introducing the gene into donor T cells given to enhance immune reconstitution in recipients of haploidentical stem-cell transplants. Patients received AP1903, an otherwise bioinert small-molecule dimerizing drug, if graft-versus-host disease (GVHD) developed. We measured the effects of AP1903 on GVHD and on the function and persistence of the cells containing the iCasp9 safety switch. RESULTS: Five patients between the ages of 3 and 17 years who had undergone stem-cell transplantation for relapsed acute leukemia were treated with the genetically modified T cells. The cells were detected in peripheral blood from all five patients and increased in number over time, despite their constitutive transgene expression. A single dose of dimerizing drug, given to four patients in whom GVHD developed, eliminated more than 90% of the modified T cells within 30 minutes after administration and ended the GVHD without recurrence. CONCLUSIONS: The iCasp9 cell-suicide system may increase the safety of cellular therapies and expand their clinical applications. (Funded by the National Heart, Lung, and Blood Institute and the National Cancer Institute; ClinicalTrials.gov number, NCT00710892.).


Subject(s)
Caspase 9/genetics , Genes, Transgenic, Suicide , Graft vs Host Disease/therapy , Immunotherapy, Adoptive , T-Lymphocytes/transplantation , Tacrolimus Binding Proteins/genetics , Adolescent , Apoptosis , Caspase 9/metabolism , Child , Child, Preschool , Female , Gene Transfer Techniques , Humans , Leukemia/therapy , Male , Organic Chemicals/therapeutic use , Recurrence , Stem Cell Transplantation , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL