Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Clin Oncol ; 35(29): 3322-3329, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28809608

ABSTRACT

Purpose Adoptive transfer of genetically modified T cells is being explored as a treatment for patients with metastatic cancer. Most current strategies use genes that encode major histocompatibility complex (MHC) class I-restricted T-cell receptors (TCRs) or chimeric antigen receptors to genetically modify CD8+ T cells or bulk T cells for treatment. Here, we evaluated the safety and efficacy of an adoptive CD4+ T-cell therapy using an MHC class II-restricted, HLA-DPB1*0401-restricted TCR that recognized the cancer germline antigen, MAGE-A3 (melanoma-associated antigen-A3). Patients and Methods Patients received a lymphodepleting preparative regimen, followed by adoptive transfer of purified CD4+ T cells, retrovirally transduced with MAGE-A3 TCR plus systemic high-dose IL-2. A cell dose escalation was conducted, starting at 107 total cells and escalating at half-log increments to approximately 1011 cells. Nine patients were treated at the highest dose level (0.78 to 1.23 × 1011 cells). Results Seventeen patients were treated. During the cell dose-escalation phase, an objective complete response was observed in a patient with metastatic cervical cancer who received 2.7 × 109 cells (ongoing at ≥ 29 months). Among nine patients who were treated at the highest dose level, objective partial responses were observed in a patient with esophageal cancer (duration, 4 months), a patient with urothelial cancer (ongoing at ≥ 19 months), and a patient with osteosarcoma (duration, 4 months). Most patients experienced transient fevers and the expected hematologic toxicities from lymphodepletion pretreatment. Two patients experienced transient grade 3 and 4 transaminase elevations. There were no treatment-related deaths. Conclusion These results demonstrate the safety and efficacy of administering autologous CD4+ T cells that are genetically engineered to express an MHC class II-restricted antitumor TCR that targets MAGE-A3. This clinical trial extends the reach of TCR gene therapy for patients with metastatic cancer.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/transplantation , Genetic Therapy/methods , HLA-DP beta-Chains/immunology , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Administration, Intravenous , Aged , Antineoplastic Agents/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Female , Genetic Therapy/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Interleukin-2/administration & dosage , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/genetics , Time Factors , Transplantation, Autologous , Treatment Outcome
2.
Mol Ther ; 25(10): 2245-2253, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28803861

ABSTRACT

T cells expressing anti-CD19 chimeric antigen receptors (CARs) can induce complete remissions (CRs) of diffuse large B cell lymphoma (DLBCL). The long-term durability of these remissions is unknown. We administered anti-CD19 CAR T cells preceded by cyclophosphamide and fludarabine conditioning chemotherapy to patients with relapsed DLBCL. Five of the seven evaluable patients obtained CRs. Four of the five CRs had long-term durability with durations of remission of 56, 51, 44, and 38 months; to date, none of these four cases of lymphomas have relapsed. Importantly, CRs continued after recovery of non-malignant polyclonal B cells in three of four patients with long-term complete remissions. In these three patients, recovery of CD19+ polyclonal B cells took place 28, 38, and 28 months prior to the last follow-up, and each of these three patients remained in CR at the last follow-up. Non-malignant CD19+ B cell recovery with continuing CRs demonstrated that remissions of DLBCL can continue after the disappearance of functionally effective anti-CD19 CAR T cell populations. Patients had a low incidence of severe infections despite long periods of B cell depletion and hypogammaglobulinemia. Only one hospitalization for an infection occurred among the four patients with long-term CRs. Anti-CD19 CAR T cells caused long-term remissions of chemotherapy-refractory DLBCL without substantial chronic toxicities.


Subject(s)
Antigens, CD19/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Receptors, Antigen, T-Cell/immunology , Adult , B-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Female , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunotherapy, Adoptive/methods , Lymphoma, Large B-Cell, Diffuse/blood , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Male , Middle Aged , T-Lymphocytes/immunology , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
3.
J Clin Oncol ; 35(16): 1803-1813, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28291388

ABSTRACT

Purpose T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 (CAR-19) have potent activity against acute lymphoblastic leukemia, but fewer results supporting treatment of lymphoma with CAR-19 T cells have been published. Patients with lymphoma that is chemotherapy refractory or relapsed after autologous stem-cell transplantation have a grim prognosis, and new treatments for these patients are clearly needed. Chemotherapy administered before adoptive T-cell transfer has been shown to enhance the antimalignancy activity of adoptively transferred T cells. Patients and Methods We treated 22 patients with advanced-stage lymphoma in a clinical trial of CAR-19 T cells preceded by low-dose chemotherapy. Nineteen patients had diffuse large B-cell lymphoma, two patients had follicular lymphoma, and one patient had mantle cell lymphoma. Patients received a single dose of CAR-19 T cells 2 days after a low-dose chemotherapy conditioning regimen of cyclophosphamide plus fludarabine. Results The overall remission rate was 73% with 55% complete remissions and 18% partial remissions. Eleven of 12 complete remissions are ongoing. Fifty-five percent of patients had grade 3 or 4 neurologic toxicities that completely resolved. The low-dose chemotherapy conditioning regimen depleted blood lymphocytes and increased serum interleukin-15 (IL-15). Patients who achieved a remission had a median peak blood CAR+ cell level of 98/µL and those who did not achieve a remission had a median peak blood CAR+ cell level of 15/µL ( P = .027). High serum IL-15 levels were associated with high peak blood CAR+ cell levels ( P = .001) and remissions of lymphoma ( P < .001). Conclusion CAR-19 T cells preceded by low-dose chemotherapy induced remission of advanced-stage lymphoma, and high serum IL-15 levels were associated with the effectiveness of this treatment regimen. CAR-19 T cells will likely become an important treatment for patients with relapsed lymphoma.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-15/blood , Lymphoma/therapy , T-Lymphocytes/transplantation , Adult , Aged , Antigens, CD19/immunology , Cyclophosphamide/administration & dosage , Humans , Interleukin-15/immunology , Lymphoma/blood , Lymphoma/drug therapy , Lymphoma/immunology , Middle Aged , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transplantation Conditioning/methods , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
4.
Hum Gene Ther Methods ; 27(6): 209-218, 2016 12.
Article in English | MEDLINE | ID: mdl-27897048

ABSTRACT

The treatment of B-cell malignancies by adoptive cell transfer (ACT) of anti-CD19 chimeric antigen receptor T cells (CD19 CAR-T) has proven to be a highly successful therapeutic modality in several clinical trials.1-6 The anti-CD19 CAR-T cell production method used to support initial trials relied on numerous manual, open process steps, human serum, and 10 days of cell culture to achieve a clinical dose.7 This approach limited the ability to support large multicenter clinical trials, as well as scale up for commercial cell production. Therefore, studies were completed to streamline and optimize the original National Cancer Institute production process by removing human serum from the process in order to minimize the risk of viral contamination, moving process steps from an open system to functionally closed system operations in order to minimize the risk of microbial contamination, and standardizing additional process steps in order to maximize process consistency. This study reports a procedure for generating CD19 CAR-T cells in 6 days, using a functionally closed manufacturing process and defined, serum-free medium. This method is able to produce CD19 CAR-T cells that are phenotypically and functionally indistinguishable from cells produced for clinical trials by the previously described production process.


Subject(s)
Antigens, CD19/immunology , Cell- and Tissue-Based Therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Clinical Trials as Topic , Culture Media, Serum-Free , Genetic Vectors/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/transplantation
5.
J Clin Oncol ; 34(10): 1112-21, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26811520

ABSTRACT

PURPOSE: Progressive malignancy is the leading cause of death after allogeneic hematopoietic stem-cell transplantation (alloHSCT). After alloHSCT, B-cell malignancies often are treated with unmanipulated donor lymphocyte infusions (DLIs) from the transplant donor. DLIs frequently are not effective at eradicating malignancy and often cause graft-versus-host disease, a potentially lethal immune response against normal recipient tissues. METHODS: We conducted a clinical trial of allogeneic T cells genetically engineered to express a chimeric antigen receptor (CAR) targeting the B-cell antigen CD19. Patients with B-cell malignancies that had progressed after alloHSCT received a single infusion of CAR T cells. No chemotherapy or other therapies were administered. The T cells were obtained from each recipient's alloHSCT donor. RESULTS: Eight of 20 treated patients obtained remission, which included six complete remissions (CRs) and two partial remissions. The response rate was highest for acute lymphoblastic leukemia, with four of five patients obtaining minimal residual disease-negative CR. Responses also occurred in chronic lymphocytic leukemia and lymphoma. The longest ongoing CR was more than 30 months in a patient with chronic lymphocytic leukemia. New-onset acute graft-versus-host disease after CAR T-cell infusion developed in none of the patients. Toxicities included fever, tachycardia, and hypotension. Peak blood CAR T-cell levels were higher in patients who obtained remissions than in those who did not. Programmed cell death protein-1 expression was significantly elevated on CAR T cells after infusion. Presence of blood B cells before CAR T-cell infusion was associated with higher postinfusion CAR T-cell levels. CONCLUSION: Allogeneic anti-CD19 CAR T cells can effectively treat B-cell malignancies that progress after alloHSCT. The findings point toward a future when antigen-specific T-cell therapies will play a central role in alloHSCT.


Subject(s)
Antigens, CD19/immunology , Hematopoietic Stem Cell Transplantation , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/surgery , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transplantation Chimera , Adult , Aged , Disease Progression , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia, B-Cell/immunology , Leukemia, B-Cell/surgery , Male , Middle Aged , Remission Induction , Transplantation, Homologous
6.
Clin Cancer Res ; 18(18): 4877-82, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22718858

ABSTRACT

Tumor-associated myeloid cells are the major type of inflammatory cells involved in the regulation of antitumor immune responses. One key characteristic of these cells is the generation of reactive oxygen (ROS) and reactive nitrogen species (RNS) in the tumor microenvironment. Recent studies have shown the important role of ROS and RNS, especially peroxynitrite, in immune suppression in cancer. ROS and RNS are involved in induction of antigen-specific T-cell tolerance, inhibition of T-cell migration to the tumor site, and tumor cell evasion of recognition by cytotoxic T cells. In preclinical settings, a number of potential therapeutic agents showed activity in blocking ROS/RNS in cancer and in improving the efficacy of cancer immune therapy. A better understanding of ROS/RNS-associated pathways in myeloid cells will help to identify more specific and direct targets to facilitate the development of more effective immune therapy for cancer.


Subject(s)
Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment/immunology , Humans , Neoplasms/pathology , Peroxynitrous Acid/metabolism , Reactive Nitrogen Species/metabolism , Translational Research, Biomedical
7.
J Clin Invest ; 121(10): 4015-29, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21911941

ABSTRACT

Cancer immunotherapeutic approaches induce tumor-specific immune responses, in particular CTL responses, in many patients treated. However, such approaches are clinically beneficial to only a few patients. We set out to investigate one possible explanation for the failure of CTLs to eliminate tumors, specifically, the concept that this failure is not dependent on inhibition of T cell function. In a previous study, we found that in mice, myeloid-derived suppressor cells (MDSCs) are a source of the free radical peroxynitrite (PNT). Here, we show that pre-treatment of mouse and human tumor cells with PNT or with MDSCs inhibits binding of processed peptides to tumor cell-associated MHC, and as a result, tumor cells become resistant to antigen-specific CTLs. This effect was abrogated in MDSCs treated with a PNT inhibitor. In a mouse model of tumor-associated inflammation in which the antitumor effects of antigen-specific CTLs are eradicated by expression of IL-1ß in the tumor cells, we determined that therapeutic failure was not caused by more profound suppression of CTLs by IL-1ß-expressing tumors than tumors not expressing this proinflammatory cytokine. Rather, therapeutic failure was a result of the presence of PNT. Clinical relevance for these data was suggested by the observation that myeloid cells were the predominant source of PNT in human lung, pancreatic, and breast cancer samples. Our data therefore suggest what we believe to be a novel mechanism of MDSC-mediated tumor cell resistance to CTLs.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/immunology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Female , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/pharmacology , Tumor Escape/drug effects , Tumor Escape/physiology
8.
J Pharmacol Exp Ther ; 319(1): 269-76, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16837556

ABSTRACT

Marijuana cannabinoids, such as delta-9-tetrahydrocannabinoid (THC), suppress type 1 T-helper 1 (Th1) immunity in a variety of models, including infection with the intracellular pathogen Legionella pneumophila (Lp). To examine the cellular mechanism of this effect, bone marrow-derived dendritic cells (DCs) were purified from BALB/c mice and studied following infection and drug treatment. DCs infected in vitro with Lp were able to protect mice when injected prior to a lethal Lp infection; however, the immunization potential of the Lp-loaded cells along with Th1 cytokine production was attenuated by THC treatment at the time of in vitro infection. In addition, THC-treated and Lp-loaded DCs were poorly stimulated in culture-primed splenic CD4(+) T cells to produce interferon-gamma; however, this stimulating deficiency was reversed by adding recombinant interleukin (IL)-12p40 protein to the cultures. Moreover, THC treatment inhibited the expression of DC maturation markers, such as major histocompatibility complex class II and costimulatory molecules CD86 and CD40 as determined by flow cytometry and suppressed the Notch ligand, Del-ta4, as determined by reverse transcription-polymerase chain reaction. However, THC treatment did not affect other DC functions, such as intracellular killing of Lp, determined by colony-forming unit counts of bacteria, and Lp-induced apoptosis, determined by annexin V staining. In conclusion, the data suggest that THC inhibits Th1 activation by targeting essential DC functions, such as IL-12p40 secretion, maturation, and expression of costimulatory and polarizing molecules.


Subject(s)
Dendritic Cells/drug effects , Dronabinol/pharmacology , Immunosuppressive Agents/pharmacology , Legionella pneumophila/immunology , Th1 Cells/immunology , Animals , Apoptosis , Cell Polarity , Cytokines/biosynthesis , Dendritic Cells/microbiology , Dendritic Cells/physiology , Immunization , Interleukin-12/biosynthesis , Interleukin-12 Subunit p40 , Interleukin-4/biosynthesis , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Protein Subunits/biosynthesis
9.
Eur J Pharmacol ; 532(1-2): 170-7, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16443217

ABSTRACT

Delta-9-tetrahydrocannabinol (THC) injection suppresses serum interleukin-12 (IL-12) levels in Legionella pneumophila-infected mice. Dendritic cells are a major producer of IL-12 and mouse, bone marrow-derived dendritic cell cultures produced high levels of the IL-12p40 following L. pneumophila infection. Treatment with THC suppressed this cytokine response in a concentration-dependent manner and the endocannabinoid, 2-arachidonoyolglycerol, less potently suppressed cytokine production. Dendritic cells expressed mRNA for cannabinoid receptor 1 (CB(1)), cannabinoid CB(2) receptor, and vanilloid receptor 1 (TRPV1) and the addition of the G(i) inhibitor, pertussis toxin, completely attenuated suppression induced by 3 and 6 muM THC but not by 10 muM THC. Furthermore, THC suppression was partially attenuated in dendritic cells from cannabinoid CB(1) receptor and CB(2) receptor knockout mice and in dendritic cells co-treated with THC and cannabinoid receptor antagonists. Cytokine suppression was not attenuated by pretreatment with the TRPV1 antagonist, capsazepine. These results suggest that THC-induced suppression of serum IL-12 is partly due to a suppression of IL-12 production by dendritic cells and that G(i) signaling and cannabinoid receptors, but not TRPV1, are involved in this suppressive effect.


Subject(s)
Dendritic Cells/drug effects , Dronabinol/pharmacology , Interleukin-12/metabolism , Legionella pneumophila/growth & development , Receptors, Cannabinoid/physiology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Hallucinogens/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pertussis Toxin/pharmacology , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/physiology , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/physiology , Receptors, Cannabinoid/genetics , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/physiology
10.
J Leukoc Biol ; 76(4): 854-61, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15258190

ABSTRACT

T helper cell type 1 (Th1)-polarizing cytokines are induced by Legionella pneumophila infection and are suppressed by pretreatment with marijuana cannabinoids (CB). Glucocorticoids and prostaglandin E2(PGE2) are also reported to suppress Th1 polarization and are induced by Delta9-tetrahydrocannabinol (THC), so their role in the suppression of polarizing cytokines was examined. Injection of L. pneumophila or THC alone into BALB/c mice induced a rapid and transient rise in serum corticosterone (CS), and the injection of both agents significantly augmented the CS response, demonstrating that THC increased CS in Legionella-infected mice. Pretreatment with the CB receptor 1 (CB1) antagonist SR141716A had no effect on the THC-induced CS response, but CB2 antagonist (SR144528) treatment increased the CS response. To see if increased CS contributed to the down-regulation of Th1 cytokines, mice were pretreated with the steroid antagonist RU486 before THC injection and Legionella infection. The results showed that RU486 did not attenuate the THC-induced suppression of serum interleukin (IL)-12 or interferon-gamma (IFN-gamma). In addition to CS, THC injection increased urinary PGE2 metabolites, and the CB1 antagonist attenuated this increase. Although L. pneumophila infection increased urinary PGE2, THC pretreatment did not enhance this response; in addition, treatment with the cyclooxygenase inhibitor, indomethacin, did not block the THC-induced suppression of IL-12 and IFN-gamma. These results suggest that the elevation of CS and PGE2 does not account for the THC-induced attenuation of the Th1 cytokine response, and it is concluded that other suppressive mediators are induced by THC or that the drug acts directly on immune cells to suppress cytokine production.


Subject(s)
Corticosterone/blood , Dinoprostone/metabolism , Dronabinol/administration & dosage , Legionella pneumophila/physiology , Legionnaires' Disease/immunology , Th1 Cells/immunology , Animals , Camphanes/pharmacology , Female , Hormone Antagonists/pharmacology , Immunosuppression Therapy , Interferon-gamma/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Radioimmunoassay , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Rimonabant
SELECTION OF CITATIONS
SEARCH DETAIL
...