Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncology ; 94(4): 223-232, 2018.
Article in English | MEDLINE | ID: mdl-29444512

ABSTRACT

OBJECTIVE: Chemotherapy with irinotecan plus cisplatin has shown promise in chemo-naïve small-cell lung cancer (SCLC) patients. However, irinotecan treatment for relapsed or refractory SCLC has not been adequately evaluated. This phase II study evaluated the appropriate treatment schedule of irinotecan as a single agent. This study was designed to determine the antitumor activity, toxicity, and survival in previously treated SCLC patients. METHODS: Previously treated SCLC patients with at least one platinum-based regimen received irinotecan (100 mg/m2) on days 1 and 8, every 3 weeks, until disease progression. The assessment of the response rate was the primary endpoint. RESULTS: Thirty patients were enrolled, with an objective response rate of 41.3% (95% confidence interval [CI] 25.5-59.3), and a disease control rate of 69%. Median progression-free and overall survival was 4.1 months (95% CI, 2.2-5.4) and 10.4 months (95% CI, 8.1-14), respectively. The grade 3/4 hematological toxicities were neutropenia (36.7%), thrombocytopenia (3.3%), anemia (13.3%), and febrile neutropenia (6.6%). There were no grade 4 nonhematological toxicities. Frequent grade 3 nonhematological toxicities included diarrhea (10%), anorexia (6.6%), and hyponatremia (6.6%). CONCLUSIONS: This phase II study showed a high objective response rate and long survival. Irinotecan monotherapy schedule used was well tolerated, and could be an active treatment option for these patients.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/analogs & derivatives , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Aged , Anorexia/chemically induced , Antineoplastic Agents, Phytogenic/adverse effects , Camptothecin/administration & dosage , Camptothecin/adverse effects , Chemotherapy-Induced Febrile Neutropenia/etiology , Diarrhea/chemically induced , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Hyponatremia/chemically induced , Irinotecan , Male , Middle Aged , Response Evaluation Criteria in Solid Tumors , Retreatment , Survival Rate , Thrombocytopenia/chemically induced
2.
PLoS One ; 12(8): e0183976, 2017.
Article in English | MEDLINE | ID: mdl-28854279

ABSTRACT

The adoptive transfer of effector T cells combined with lymphodepletion has demonstrated promising antitumor effects in mice and humans, although the availability of tumor-specific T cells is limited. We and others have also demonstrated that the transfer of polyclonal naïve T cells induces tumor-specific effector T cells and enhances antitumor immunity after lymphodepletion. Because tumors have been demonstrated to induce immunosuppressive networks and regulate the function of T cells, obtaining a sufficient number of fully functional naïve T cells that are able to differentiate into tumor-specific effector T cells remains difficult. To establish culture methods to obtain a large number of polyclonal T cells that are capable of differentiating into tumor-specific effector T cells, naïve T cells were activated with anti-CD3 mAbs in vitro. These cells were stimulated with IL-2 and IL-7 for the CD8 subset or with IL-7 and IL-23 for the CD4 subset. Transfer of these hyperexpanded T cells after lymphodepletion showed significant antitumor efficacy, and tumor-specific effector T cells were primed from these expanded T cells in tumor-bearing hosts. Moreover, these ex vivo-expanded T cells maintained T cell receptor diversity and showed long-term persistence of memory against specific tumors. Further analyses revealed that combination therapy consisting of vaccination with dendritic cells that were co-cultured with irradiated whole tumor cells and the transfer of ex vivo-expanded T cells significantly enhanced antitumor immunity. These results indicate that the transfer of ex vivo-expanded polyclonal T cells can be combined with other immunotherapies and augment antitumor effects.


Subject(s)
Adoptive Transfer/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/transplantation , Female , Interleukin-2/immunology , Interleukin-23/immunology , Interleukin-7/immunology , Lymphocyte Activation , Lymphocyte Depletion , Mice, Inbred C57BL , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology
3.
J Immunol ; 195(2): 726-35, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26041539

ABSTRACT

Antitumor immunity is augmented by cytotoxic lymphodepletion therapies. Adoptively transferred naive and effector T cells proliferate extensively and show enhanced antitumor effects in lymphopenic recipients. Although the impact of lymphodepletion on transferred donor T cells has been well evaluated, its influence on recipient T cells is largely unknown. The current study demonstrates that both regulatory T cells (Tregs) and effector CD8(+) T cells from lymphopenic recipients play critical roles in the development of antitumor immunity after lymphodepletion. Cyclophosphamide (CPA) treatment depleted lymphocytes more efficiently than other cytotoxic agents; however, the percentage of CD4(+)CD25(+) Foxp3(+) Tregs was significantly increased in CPA-treated lymphopenic mice. Depletion of these chemoresistant Tregs following CPA treatment and transfer of naive CD4(+) T cells augmented the antitumor immunity and significantly suppressed tumor progression. Further analyses revealed that recipient CD8(+) T cells were responsible for this augmentation. Using Rag2(-/-) mice or depletion of recipient CD8(+) T cells after CPA treatment abrogated the augmentation of antitumor effects in CPA-treated reconstituted mice. The transfer of donor CD4(+) T cells enhanced the proliferation of CD8(+) T cells and the priming of tumor-specific CD8(+) T cells originating from the lymphopenic recipients. These results highlight the importance of the recipient cells surviving cytotoxic regimens in cancer immunotherapies.


Subject(s)
Cytotoxins/pharmacology , Fibrosarcoma/therapy , Lymphocyte Depletion , Lymphopenia/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cisplatin/pharmacology , Cyclophosphamide/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance/immunology , Etoposide/pharmacology , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphopenia/chemically induced , Lymphopenia/immunology , Lymphopenia/pathology , Methylcholanthrene , Mice , Mice, Inbred C57BL , Mice, Knockout , Paclitaxel/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Tumor Cells, Cultured , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Whole-Body Irradiation , Gemcitabine
4.
Blood ; 120(12): 2417-27, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22806892

ABSTRACT

Cytotoxic lymphodepletion therapies augment antitumor immune responses. The generation and therapeutic efficacy of antitumor effector T cells (T(E)s) are enhanced during recovery from lymphopenia. Although the effects of lymphodepletion on naive T cells (T(N)s) and T(E)s have been studied extensively, the influence of lymphodepletion on suppressor cells remains poorly understood. In this study, we demonstrate a significant increase of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) in sublethally irradiated lymphopenic mice. These radio-resistant Tregs inhibited the induction of T(E)s in tumor-draining lymph-nodes (TDLNs) during recovery from lymphopenia. The transfer of T(N)s into lymphopenic tumor-bearing mice resulted in some antitumor effects; however, Treg depletion after whole-body irradiation and reconstitution strongly inhibited tumor progression. Further analyses revealed that tumor-specific T cells were primed from the transferred T(N)s, whereas the Tregs originated from irradiated recipient cells. As in irradiated lymphopenic mice, a high percentage of Tregs was observed in cyclophosphamide-treated lymphopenic mice. The inhibition of Tregs in cyclophosphamide-treated mice significantly reduced tumor growth. These results indicate that the Tregs that survive cytotoxic therapies suppress antitumor immunity during recovery from lymphopenia and suggest that approaches to deplete radio and chemo-resistant Tregs can enhance cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Depletion , Lymphopenia/immunology , Lymphopenia/therapy , Radiation Injuries, Experimental/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Cell Proliferation , Combined Modality Therapy , Cyclophosphamide/therapeutic use , Flow Cytometry , Forkhead Transcription Factors/metabolism , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/pathology , Whole-Body Irradiation
5.
Cancer Immunol Immunother ; 60(11): 1597-608, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21691723

ABSTRACT

Accumulating evidence suggests that cancer cells possess a small subpopulation that survives during potentially lethal stresses, including chemotherapy, radiation treatment, and molecular-targeting therapy. CD133 is a putative marker that distinguishes a minor subpopulation from normal differentiated tumor cells in many cancers. Although it is necessary to eradicate all cancer cells to obtain a cure, effective treatment to eliminate the CD133(+) treatment-tolerant cells has not been elucidated. In this study, we demonstrated that a CD133(+) subpopulation in murine melanoma is immunogenic and that effector T cells specific for the CD133(+) melanoma cells mediated potent antitumor reactivity, curing the mice of the parental melanoma. CD133(+) melanoma antigens preferentially induced type 17 T helper (Th17) cells and Th1 cells but not Th2 cells. CD133(+) melanoma cell-specific CD4(+) T-cell treatment eradicated not only CD133(+) tumor cells but also CD133(-) tumor cells while inducing long-lasting accumulation of lymphocytes and dendritic cells with upregulated MHC class II in tumor tissues. Further, the treatment prevented regulatory T-cell induction. These results indicate that T-cell immunotherapy is a promising treatment option to eradicate CD133(+) drug-tolerant cells to obtain a cure for cancer.


Subject(s)
Antigens, CD/immunology , Glycoproteins/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Peptides/immunology , T-Lymphocytes/immunology , Th1 Cells/immunology , Th17 Cells/immunology , AC133 Antigen , Animals , Antigens, CD/pharmacology , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Glycoproteins/pharmacology , Immunotherapy, Active/methods , Immunotherapy, Adoptive/methods , Major Histocompatibility Complex , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Peptides/pharmacology , T-Lymphocytes/pathology , Th1 Cells/pathology , Th17 Cells/pathology
6.
BMC Cancer ; 11: 1, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21194487

ABSTRACT

BACKGROUND: Gefitinib was the first epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) approved for the treatment of advanced non-small cell lung cancer (NSCLC). Few treatment options are available for NSCLC patients who have responded to gefitinib treatment and demonstrated tumor progression. The present study was conducted to evaluate the efficacy and toxicity of the 2(nd) EGFR-TKI administration. METHODS: We retrospectively analyzed 11 patients who had obtained a partial response (PR) or stable disease (SD) with gefitinib treatment and were re-treated with EGFR-TKI after failure of the initial gefitinib treatment. RESULTS: Three patients (27%) were treated with gefitinib as the 2(nd) EGFR-TKI, and 8 patients (73%) received erlotinib. Only one patient (9%) showed PR, 7 (64%) achieved SD, and 3 (27%) had progressive disease. The disease control rate was 73% (95% CI, 43% - 91%) and the median progression-free survival was 3.4 months (95% CI, 2 - 5.2). The median overall survival from the beginning of the 2(nd) EGFR-TKI and from diagnosis were 7.3 months (95% CI, 2.7 - 13) and 36.7 months (95% CI, 23.6 - 43.9), respectively. No statistical differences in PFS or OS were observed between gefitinib and erlotinib as the 2(nd) EGFR-TKI (PFS, P = 0.23 and OS, P = 0.052). The toxicities associated with the 2(nd) EGFR-TKI were generally acceptable and comparable to those observed for the initial gefitinib therapy. CONCLUSIONS: Our results indicate that a 2(nd) EGFR-TKI treatment can be an effective treatment option for gefitinib responders.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Aged , Diarrhea/chemically induced , Erlotinib Hydrochloride , Exanthema/chemically induced , Female , Gefitinib , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Quinazolines/adverse effects , Retreatment , Retrospective Studies , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...