Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 204
Filter
Add more filters










Publication year range
2.
J Immunother Cancer ; 7(1): 216, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409394

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) for solid tumors, including those targeting programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), have shown impressive clinical efficacy, however, most patients do not achieve durable responses. One major therapeutic obstacle is the immunosuppressive tumor immune microenvironment (TIME). Thus, we hypothesized that a strategy combining tumor-directed radiation with TIME immunomodulation could improve ICI response rates in established solid tumors. METHODS: Using a syngeneic mouse model of human papillomavirus (HPV)-associated head and neck cancer, mEER, we developed a maximally effective regimen combining PD-1 and CTLA-4 inhibition, tumor-directed radiation, and two existing immunomodulatory drugs: cyclophosphamide (CTX) and a small-molecule inducible nitric oxide synthase (iNOS) inhibitor, L-n6-(1-iminoethyl)-lysine (L-NIL). We compared the effects of the various combinations of this regimen on tumor growth, overall survival, establishment of immunologic memory, and immunologic changes with flow cytometry and quantitative multiplex immunofluorescence. RESULTS: We found PD-1 and CTLA-4 blockade, and radiotherapy alone or in combination, incapable of clearing established tumors or reversing the unfavorable balance of effector to suppressor cells in the TIME. However, modulation of the TIME with cyclophosphamide (CTX) and L-NIL in combination with dual checkpoint inhibition and radiation led to rejection of over 70% of established mEER tumors and doubled median survival in the B16 melanoma model. Anti-tumor activity was CD8+ T cell-dependent and led to development of immunologic memory against tumor-associated HPV antigens. Immune profiling revealed that CTX/L-NIL induced remodeling of myeloid cell populations in the TIME and tumor-draining lymph node and drove subsequent activation and intratumoral infiltration of CD8+ effector T cells. CONCLUSIONS: Overall, this study demonstrates that modulation of the immunosuppressive TIME is required to unlock the benefits of ICIs and radiotherapy to induce immunologic rejection of treatment-refractory established solid tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Humans , Male , Mice , Tumor Microenvironment
3.
J Immunother Cancer ; 7(1): 10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30646957

ABSTRACT

BACKGROUND: Chemoradiotherapy (CRT) remains one of the most common cancer treatment modalities, and recent data suggest that CRT is maximally effective when there is generation of an anti-tumoral immune response. However, CRT has also been shown to promote immunosuppressive mechanisms which must be blocked or reversed to maximize its immune stimulating effects. METHODS: Therefore, using a preclinical model of human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC), we developed a clinically relevant therapy combining CRT and two existing immunomodulatory drugs: cyclophosphamide (CTX) and the small molecule inducible nitric oxide synthase (iNOS) inhibitor L-n6-(1-iminoethyl)-lysine (L-NIL). In this model, we treated the syngeneic HPV-HNSCC mEER tumor-bearing mice with fractionated (10 fractions of 3 Gy) tumor-directed radiation and weekly cisplatin administration. We compared the immune responses induced by CRT and those induced by combinatory treatment (CRT + CTX/L-NIL) with flow cytometry, quantitative multiplex immunofluorescence and by profiling immune-related gene expression changes. RESULTS: We show that combination treatment favorably remodels the tumor myeloid immune microenvironment including an increase in anti-tumor immune cell types (inflammatory monocytes and M1-like macrophages) and a decrease in immunosuppressive granulocytic myeloid-derived suppressor cells (MDSCs). Intratumoral T cell infiltration and tumor antigen specificity of T cells were also improved, including a 31.8-fold increase in the CD8+ T cell/ regulatory T cell ratio and a significant increase in tumor antigen-specific CD8+ T cells compared to CRT alone. CTX/LNIL immunomodulation was also shown to significantly improve CRT efficacy, leading to rejection of 21% established tumors in a CD8-dependent manner. CONCLUSIONS: Overall, these data show that modulation of the tumor immune microenvironment with CTX/L-NIL enhances susceptibility of treatment-refractory tumors to CRT. The combination of tumor immune microenvironment modulation with CRT constitutes a translationally relevant approach to enhance CRT efficacy through enhanced immune activation.


Subject(s)
Antineoplastic Agents/therapeutic use , Chemoradiotherapy , Cyclophosphamide/therapeutic use , Head and Neck Neoplasms/therapy , Immunomodulation , Lysine/analogs & derivatives , Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Head and Neck Neoplasms/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lysine/therapeutic use , Male , Mice, Inbred C57BL , Papillomavirus Infections/complications , Squamous Cell Carcinoma of Head and Neck/immunology
4.
Oncoimmunology ; 7(10): e1490853, 2018.
Article in English | MEDLINE | ID: mdl-30288358

ABSTRACT

Cancer-induced myeloid-derived suppressor cells (MDSC) play an important role in tumor immune evasion. MDSC programming or polarization has been proposed as a strategy for leveraging the developmental plasticity of myeloid cells to reverse MDSC immune suppressive functions, or cause them to acquire anti-tumor activity. While MDSC derived ex vivo from murine bone marrow precursor cells with tumor-conditioned medium efficiently suppressed T cell proliferation, MDSC derived from conditioned medium in presence of TGF-ß1 (TGFß-MDSC) acquired a novel immune-stimulatory phenotype, losing the ability to inhibit T cell proliferation and acquiring enhanced antigen-presenting capability. Altered immune function was associated with SMAD-2 dependent upregulation of maturation and costimulatory molecules, and downregulation of inducible nitric oxide synthase (iNOS), an effector mechanism of immunosuppression. TGFß-MDSC also upregulated FAS-ligand expression, leading to FAS-dependent killing of murine human papillomavirus (HPV)-associated head and neck cancer cells and tumor spheroids in vitro and anti-tumor activity in vivo. Radiation upregulated FAS expression on tumor cells, and the combination of radiotherapy and intratumoral injection of TGFß-MDSC strongly enhanced class I expression on tumor cells and induction of HPV E7 tetramer-positive CD8 + T cells, leading to clearance of established tumors and long-term survival. TGFß-MDSC derived from human PBMC with tumor conditioned medium also lost immunosuppressive function and acquired tumor-killing activity. Thus, TGFß1 mediated programming of nascent MDSC leads to a potent anti-tumor phenotype potentially suitable for adoptive immunotherapy.

5.
Cancer Res ; 78(21): 6159-6170, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30154146

ABSTRACT

Human papillomavirus subtype 16 (HPV16) is the primary cause of an increasing number of head and neck squamous cell carcinomas (HNSCC), providing strong rationale for T-cell immune therapies against HPV+ HNSCC. Here we assess immunogenicity of HPV16-specific CD8+ T cells (CTL) and characterize HPV-specific mechanisms of T-cell dysfunction. We identified 16 strong and 29 moderately immunogenic CTL-epitopes from HPV16 E2, E6, and E7 antigens restricted by 12 common HLA class I alleles. E2-specific CTL-reactivity was higher in patients with HPV+ HNSCC than in healthy controls (>3-fold; P = 0.026). Patient-derived E2, E6, and E7 peripheral CTLs exhibited heterogeneity in dysfunctional phenotypes. Immunogenomic analyses of 119 HNSCC transcriptomes revealed high T-cell infiltration and dysfunction in HPV+ HNSCC and correlation of HPV antigen expression with T-cell exhaustion gene signatures. Indoleamine 2,3-dioxygenase (IDO-1) was strongly expressed in HPV+ HNSCC versus HPV- HNSCC (P = 0.001) and correlated with E7 expression (R 2 = 0.84; P = 0.033). Combination treatment with PD-1 blockade and IDO-1 inhibition overcame profound CTL-dysfunction, enhancing HPV+ HNSCC sensitivity to CTL-cytotoxicity in vitro (up to 10-fold in E7-CTLs, P = 0.011). Our findings implicate mechanisms of T-cell escape in HPV+ HNSCC, wherein high tumoral HPV-antigen load results in high expression of immune dysfunction genes on tumor cells (e.g., IDO-1), and dysfunction of HPV-specific CTLs (e.g., E7, E2-CTLs). The HPV16 CTL-epitopes identified in this study, in combination with blockade of HPV+ HNSCC-specific PD-1/IDO-1 checkpoints, may be useful for targeted immunotherapy.Significance: This study evaluates the HPV antigen T-cell immunogenicity role of inhibitory receptors and other exhaustion markers in the cytotoxic function of HPV antigen-specific CTLs and identifies combined inhibition of PD-1/IDO-1 as a strategy to enhance CTL targeting of HPV+ HNSCC. Cancer Res; 78(21); 6159-70. ©2018 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Head and Neck Neoplasms/metabolism , Human papillomavirus 16/immunology , Aged , Alleles , Epitope Mapping , Epitopes/immunology , Head and Neck Neoplasms/virology , Histocompatibility Antigens Class I/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Leukocytes, Mononuclear/cytology , Middle Aged , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Phenotype , T-Lymphocytes, Cytotoxic/cytology
8.
J Assoc Physicians India ; 64(2): 11-12, 2016 02.
Article in English | MEDLINE | ID: mdl-27730774

ABSTRACT

Febrile patient with thrombocytopenia is commonly encountered by physicians especially during monsoon and perimonsoon period. Infections with protozoa, bacteria and viruses can cause thrombocytopenia with or without disseminated intravascular coagulation. Commonly dengue, malaria, scrub typhus and other rickettsial infections, meningococci, leptospira and certain viral infections present as fever with thrombocytopenia. Occasionally these patients can go on to develop a stormy course with multiorgan dysfunction requiring intensive care unit admission associated with high morbidity and mortality.1,2 Infections cause decrease in platelet count both due to effects on platelet production and platelet survival.3 Thrombocytopenia in bacterial infections can occur as a part of sepsis with disseminated intravascular coagulation. Patients with sepsis may also develop hemophagocytic histiocytosis with phagocytosis of platelets and leucocytes in the bone marrow histiocytes. Both Gram-positive and Gram-negative bacterial infections can lead to sepsis. Elevated platelet-associated IgG has been implicated. Platelets tend to adhere to damaged vascular surfaces in meningococcemia.


Subject(s)
Bacterial Infections/complications , Blood Platelets/pathology , Disseminated Intravascular Coagulation/complications , Sepsis/complications , Thrombocytopenia/etiology , Bacteria/pathogenicity , Disseminated Intravascular Coagulation/pathology , Humans , Platelet Count , Sepsis/metabolism
9.
Cancer Immunol Res ; 4(10): 823-834, 2016 10.
Article in English | MEDLINE | ID: mdl-27485136

ABSTRACT

Human papillomavirus (HPV), particularly HPV16 and HPV18, can cause cancers in diverse anatomical sites, including the anogenital and oropharyngeal (throat) regions. Therefore, development of safe and clinically effective therapeutic vaccines is an important goal. Herein, we show that a recombinant fusion protein of a humanized antibody to CD40 fused to HPV16.E6/7 (αCD40-HPV16.E6/7) can evoke HPV16.E6/7-specific CD8+ and CD4+ T-cell responses in head-and-neck cancer patients in vitro and in human CD40 transgenic (hCD40Tg) mice in vivo The combination of αCD40-HPV16.E6/7 and poly(I:C) efficiently primed HPV16.E6/7-specific T cells, particularly CD8+ T cells, in hCD40Tg mice. Inclusion of montanide enhanced HPV16.E6/7-specific CD4+, but not CD8+, T-cell responses. Poly(I:C) plus αCD40-HPV16.E6/7 was sufficient to mount both preventative and therapeutic immunity against TC-1 tumors in hCD40Tg mice, significantly increasing the frequency of HPV16-specific CD8+ CTLs in the tumors, but not in peripheral blood. In line with this, tumor volume inversely correlated with the frequency of HPV16.E6/7-specific CD8+ T cells in tumors, but not in blood. These data suggest that CD40-targeting vaccines for HPV-associated malignancies can provide a highly immunogenic platform with a strong likelihood of clinical benefit. Data from this study offer strong support for the development of CD40-targeting vaccines for other cancers in the future. Cancer Immunol Res; 4(10); 823-34. ©2016 AACR.


Subject(s)
CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Head and Neck Neoplasms/immunology , Papillomavirus Vaccines/immunology , Animals , Antiviral Agents/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Human papillomavirus 16/immunology , Humans , Immunity, Cellular , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/immunology , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...