Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alfonso R. Sánchez-Paulete is active.

Publication


Featured researches published by Alfonso R. Sánchez-Paulete.


Cancer Discovery | 2016

Cancer Immunotherapy with Immunomodulatory Anti-CD137 and Anti–PD-1 Monoclonal Antibodies Requires BATF3-Dependent Dendritic Cells

Alfonso R. Sánchez-Paulete; Francisco J. Cueto; María Martínez-López; Sara Labiano; Aizea Morales-Kastresana; Maria E. Rodriguez-Ruiz; Maria Jure-Kunkel; Arantza Azpilikueta; María Ángela Aznar; Jose I. Quetglas; David Sancho; Ignacio Melero

UNLABELLED Weak and ineffective antitumor cytotoxic T lymphocyte (CTL) responses can be rescued by immunomodulatory mAbs targeting PD-1 or CD137. Using Batf3(-/-) mice, which are defective for cross-presentation of cell-associated antigens, we show that BATF3-dependent dendritic cells (DC) are essential for the response to therapy with anti-CD137 or anti-PD-1 mAbs. Batf3(-/-) mice failed to prime an endogenous CTL-mediated immune response toward tumor-associated antigens, including neoantigens. As a result, the immunomodulatory mAbs could not amplify any therapeutically functional immune response in these mice. Moreover, administration of systemic sFLT3L and local poly-ICLC enhanced DC-mediated cross-priming and synergized with anti-CD137- and anti-PD-1-mediated immunostimulation in tumor therapy against B16-ovalbumin-derived melanomas, whereas this function was lost in Batf3(-/-) mice. These experiments show that cross-priming of tumor antigens by FLT3L- and BATF3-dependent DCs is crucial to the efficacy of immunostimulatory mAbs and represents a very attractive point of intervention to enhance their clinical antitumor effects. SIGNIFICANCE Immunotherapy with immunostimulatory mAbs is currently achieving durable clinical responses in different types of cancer. We show that cross-priming of tumor antigens by BATF3-dependent DCs is a key limiting factor that can be exploited to enhance the antitumor efficacy of anti-PD-1 and anti-CD137 immunostimulatory mAbs.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Focusing and sustaining the antitumor CTL effector killer response by agonist anti-CD137 mAb

Bettina Weigelin; Elixabet Bolaños; Alvaro Teijeira; Ivan Martinez-Forero; Sara Labiano; Arantza Azpilikueta; Aizea Morales-Kastresana; Jose I. Quetglas; Esther Wagena; Alfonso R. Sánchez-Paulete; Lieping Chen; Peter Friedl; Ignacio Melero

Significance Immunotherapy of cancer with immunomodulatory agents is achieving significant efficacy in an important fraction of patients. The stimulatory inducible receptor of T and NK lymphocytes known as CD137 or 4-1BB is being stimulated with agonist antibodies to enhance antitumor immunity in clinical trials. In addition, the intracellular signaling domain of CD137 is crucial as a component of successful anti-leukemia therapies with chimeric antigen receptors transduced into adoptively transferred T lymphocytes. In this study the marked synergistic effects of adoptive T cell and agonist anti-CD137 mAb therapies are studied, providing in vivo evidence for improved, more sustained and focused tumoricidal functions of antitumor cytotoxic T lymphocytes when under the influence of CD137-targeted pharmacological stimulation with immunostimulatory monoclonal antibodies. Cancer immunotherapy is undergoing significant progress due to recent clinical successes by refined adoptive T-cell transfer and immunostimulatory monoclonal Ab (mAbs). B16F10-derived OVA-expressing mouse melanomas resist curative immunotherapy with either adoptive transfer of activated anti-OVA OT1 CTLs or agonist anti-CD137 (4-1BB) mAb. However, when acting in synergistic combination, these treatments consistently achieve tumor eradication. Tumor-infiltrating lymphocytes that accomplish tumor rejection exhibit enhanced effector functions in both transferred OT-1 and endogenous cytotoxic T lymphocytes (CTLs). This is consistent with higher levels of expression of eomesodermin in transferred and endogenous CTLs and with intravital live-cell two-photon microscopy evidence for more efficacious CTL-mediated tumor cell killing. Anti-CD137 mAb treatment resulted in prolonged intratumor persistence of the OT1 CTL-effector cells and improved function with focused and confined interaction kinetics of OT-1 CTL with target cells and increased apoptosis induction lasting up to six days postadoptive transfer. The synergy of adoptive T-cell therapy and agonist anti-CD137 mAb thus results from in vivo enhancement and sustainment of effector functions.


Cancer Research | 2016

Abscopal Effects of Radiotherapy Are Enhanced by Combined Immunostimulatory mAbs and Are Dependent on CD8 T Cells and Crosspriming

Maria E. Rodriguez-Ruiz; Inmaculada Rodriguez; Saray Garasa; Benigno Barbés; Jose Luis Solorzano; Jose Luis Perez-Gracia; Sara Labiano; Miguel F. Sanmamed; Arantza Azpilikueta; Elixabet Bolaños; Alfonso R. Sánchez-Paulete; M. Angela Aznar; Ana Rouzaut; Kurt A. Schalper; Maria Jure-Kunkel; Ignacio Melero

Preclinical and clinical evidence indicate that the proimmune effects of radiotherapy can be synergistically augmented with immunostimulatory mAbs to act both on irradiated tumor lesions and on distant, nonirradiated tumor sites. The combination of radiotherapy with immunostimulatory anti-PD1 and anti-CD137 mAbs was conducive to favorable effects on distant nonirradiated tumor lesions as observed in transplanted MC38 (colorectal cancer), B16OVA (melanoma), and 4T1 (breast cancer) models. The therapeutic activity was crucially performed by CD8 T cells, as found in selective depletion experiments. Moreover, the integrities of BATF-3-dependent dendritic cells specialized in crosspresentation/crosspriming of antigens to CD8+ T cells and of the type I IFN system were absolute requirements for the antitumor effects to occur. The irradiation regimen induced immune infiltrate changes in the irradiated and nonirradiated lesions featured by reductions in the total content of effector T cells, Tregs, and myeloid-derived suppressor cells, while effector T cells expressed more intracellular IFNγ in both the irradiated and contralateral tumors. Importantly, 48 hours after irradiation, CD8+ TILs showed brighter expression of CD137 and PD1, thereby displaying more target molecules for the corresponding mAbs. Likewise, PD1 and CD137 were induced on tumor-infiltrating lymphocytes from surgically excised human carcinomas that were irradiated ex vivo These mechanisms involving crosspriming and CD8 T cells advocate clinical development of immunotherapy combinations with anti-PD1 plus anti-CD137 mAbs that can be synergistically accompanied by radiotherapy strategies, even if the disease is left outside the field of irradiation. Cancer Res; 76(20); 5994-6005. ©2016 AACR.


European Journal of Immunology | 2016

Deciphering CD137 (4‐1BB) signaling in T‐cell costimulation for translation into successful cancer immunotherapy

Alfonso R. Sánchez-Paulete; Sara Labiano; Maria E. Rodriguez-Ruiz; Arantza Azpilikueta; Iñaki Etxeberria; Elixabet Bolaños; Valérie Lang; Manuel Rodríguez; M. Angela Aznar; Maria Jure-Kunkel; Ignacio Melero

CD137 (4‐1BB, TNF‐receptor superfamily 9) is a surface glycoprotein of the TNFR family which can be induced on a variety of leukocyte subsets. On T and NK cells, CD137 is expressed following activation and, if ligated by its natural ligand (CD137L), conveys polyubiquitination‐mediated signals via TNF receptor associated factor 2 that inhibit apoptosis, while enhancing proliferation and effector functions. CD137 thus behaves as a bona fide inducible costimulatory molecule. These functional properties of CD137 can be exploited in cancer immunotherapy by systemic administration of agonist monoclonal antibodies, which increase anticancer CTLs and enhance NK‐cell‐mediated antibody‐dependent cell‐mediated cytotoxicity. Reportedly, anti‐CD137 mAb and adoptive T‐cell therapy strongly synergize, since (i) CD137 expression can be used to select the T cells endowed with the best activities against the tumor, (ii) costimulation of the lymphocyte cultures to be used in adoptive T‐cell therapy can be done with CD137 agonist antibodies or CD137L, and (iii) synergistic effects upon coadministration of T cells and antibodies are readily observed in mouse models. Furthermore, the signaling cytoplasmic tail of CD137 is a key component of anti‐CD19 chimeric antigen receptors that are used to redirect T cells against leukemia and lymphoma in the clinic. Ongoing phase II clinical trials with agonist antibodies and the presence of CD137 sequence in these successful chimeric antigen receptors highlight the importance of CD137 in oncoimmunology.


Cancer immunology research | 2015

Virotherapy with a Semliki Forest Virus–Based Vector Encoding IL12 Synergizes with PD-1/PD-L1 Blockade

Jose I. Quetglas; Sara Labiano; M. Angela Aznar; Elixabet Bolaños; Arantza Azpilikueta; Inmaculada Rodriguez; Erkuden Casales; Alfonso R. Sánchez-Paulete; Victor Segura; Cristian Smerdou; Ignacio Melero

Quetglas and colleagues report that intratumoral injection of cytolytic nonreplicative Semliki Forest virus vector expressing IL12, along with systemic administration of anti-PD-1/PD-L1 antibodies, induced regression of both virally injected and distal tumors and synergistically prolonged survival in mouse tumor models. Virotherapy and checkpoint inhibitors can be combined for the treatment of cancer with complementarity and potential for synergistic effects. We have developed a cytolytic but nonreplicative viral vector system based on Semliki Forest virus that encodes IL12 (SFV-IL12). Following direct intratumoral injection, infected cells release transgenic IL12, die, and elicit an inflammatory response triggered by both abundantly copied viral RNA and IL12. In difficult-to-treat mouse cancer models, such as those derived from MC38 and bilateral B16-OVA, SFV-IL12 synergized with an anti–PD-1 monoclonal antibody (mAb) to induce tumor regression and prolong survival. Similar synergistic effects were attained upon PD-L1 blockade. Combined SFV-IL12 + anti–PD-1 mAb treatment only marginally increased the elicited cytotoxic T-lymphocyte response over SFV-IL12 as a single agent, at least when measured by in vivo killing assays. In contrast, we observed that SFV-IL12 treatment induced expression of PD-L1 on tumor cells in an IFNγ-dependent fashion. PD-L1–mediated adaptive resistance thereby provides a mechanistic explanation of the observed synergistic effects achieved by the SFV-IL12 + anti–PD-1 mAb combination. Cancer Immunol Res; 3(5); 449–54. ©2015 AACR.


Journal of Immunology | 2017

Intratumoral Delivery of Immunotherapy—Act Locally, Think Globally

M. Angela Aznar; Nicola Tinari; Antonio J. Rullán; Alfonso R. Sánchez-Paulete; Maria E. Rodriguez-Ruiz; Ignacio Melero

Immune mechanisms have evolved to cope with local entry of microbes acting in a confined fashion but eventually inducing systemic immune memory. Indeed, in situ delivery of a number of agents into tumors can mimic in the malignant tissue the phenomena that control intracellular infection leading to the killing of infected cells. Vascular endothelium activation and lymphocyte attraction, together with dendritic cell–mediated cross-priming, are the key elements. Intratumoral therapy with pathogen-associated molecular patterns or recombinant viruses is being tested in the clinic. Cell therapies can be also delivered intratumorally, including infusion of autologous dendritic cells and even tumor-reactive T lymphocytes. Intralesional virotherapy with an HSV vector expressing GM-CSF has been recently approved by the Food and Drug Administration for the treatment of unresectable melanoma. Immunomodulatory monoclonal Abs have also been successfully applied intratumorally in animal models. Local delivery means less systemic toxicity while focusing the immune response on the malignancy and the affected draining lymph nodes.


Journal of Thoracic Oncology | 2016

Successful Immunotherapy against a Transplantable Mouse Squamous Lung Carcinoma with Anti–PD-1 and Anti-CD137 Monoclonal Antibodies

Arantza Azpilikueta; Jackeline Agorreta; Sara Labiano; Jose Luis Perez-Gracia; Alfonso R. Sánchez-Paulete; M. Angela Aznar; Daniel Ajona; Ignacio Gil-Bazo; Marta Larrayoz; Alvaro Teijeira; Maria E. Rodriguez-Ruiz; Ruben Pio; Luis M. Montuenga; Ignacio Melero

Introduction: Anti–programmed cell death 1 (anti–PD‐1) and anti–programmed cell death ligand 1 (PD‐L1) antagonist monoclonal antibodies (mAbs) against metastatic non–small cell lung cancer with special efficacy in patients with squamous cell lung cancer are being developed in the clinic. However, robust and reliable experimental models to test immunotherapeutic combinations in squamous lung tumors are still lacking. Methods: We generated a transplantable squamous cell carcinoma cell line (UN‐SCC680AJ) from a lung tumor induced by chronic N‐nitroso‐tris‐chloroethylurea mutagenesis in A/J mice. Tumor cells expressed cytokeratins, overexpressed p40, and lacked thyroid transcription factor 1, confirming the squamous lineage reported by histological analysis. More than 200 mutations found in its exome suggested potential for antigenicity. Immunocompetent mice subcutaneously implanted with this syngeneic cell line were treated with anti‐CD137 and/or anti–PD‐1 mAbs and monitored for tumor growth/progression or assessed for intratumoral leukocyte infiltration using immunohistochemical analysis and flow cytometry. Results: In syngeneic mice, large 12‐day–established tumors derived from the transplantable cell line variant UN‐SCC680AJ were amenable to curative treatment with anti–PD‐1, anti–PD‐L1, or anti‐CD137 immunostimulatory mAbs. Single‐agent therapies lost curative efficacy when treatment was started beyond day +17, whereas a combination of anti–PD‐1 plus anti‐CD137 achieved complete rejections. Tumor cells expressed weak baseline PD‐L1 on the plasma membrane, but this could be readily induced by interferon‐&ggr;. Combined treatment efficacy required CD8 T cells and induced a leukocyte infiltrate in which T lymphocytes co‐expressing CD137 and PD‐1 were prominent. Conclusions: These promising results advocate the use of combined anti–PD‐1/PD‐L1 plus anti‐CD137 mAb immunotherapy for the treatment of squamous non–small cell lung cancer in the clinical setting.


OncoImmunology | 2016

Hypoxia-induced soluble CD137 in malignant cells blocks CD137L-costimulation as an immune escape mechanism.

Sara Labiano; Asis Palazon; Elixabet Bolaños; Arantza Azpilikueta; Alfonso R. Sánchez-Paulete; Aizea Morales-Kastresana; Jose I. Quetglas; Jose Luis Perez-Gracia; Alfonso Gurpide; Maria E. Rodriguez-Ruiz; M. Angela Aznar; Maria Jure-Kunkel; Pedro Berraondo; Ignacio Melero

ABSTRACT Hypoxia is a common feature in solid tumors that has been implicated in immune evasion. Previous studies from our group have shown that hypoxia upregulates the co-stimulatory receptor CD137 on activated T lymphocytes and on vascular endothelial cells. In this study, we show that exposure of mouse and human tumor cell lines to hypoxic conditions (1% O2) promotes CD137 transcription. However, the resulting mRNA is predominantly an alternatively spliced form that encodes for a soluble variant, lacking the transmembrane domain. Accordingly, soluble CD137 (sCD137) is detectable by ELISA in the supernatant of hypoxia-exposed cell lines and in the serum of tumor-bearing mice. sCD137, as secreted by tumor cells, is able to bind to CD137-Ligand (CD137L). Our studies on primed T lymphocytes in co-culture with stable transfectants for CD137L demonstrate that tumor-secreted sCD137 prevents co-stimulation of T lymphocytes. Such an effect results from preventing the interaction of CD137L with the transmembrane forms of CD137 expressed on T lymphocytes undergoing activation. Indeed, silencing CD137 with shRNA renders more immunogenic tumor-cell variants upon inoculation to immunocompetent mice but which readily grafted on immunodeficient or CD8+ T-cell-depleted mice. These mechanisms are interpreted as a molecular strategy deployed by tumors to repress lymphocyte co-stimulation via CD137/CD137L.


Annals of Oncology | 2017

Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy

Alfonso R. Sánchez-Paulete; Alvaro Teijeira; Francisco J. Cueto; Saray Garasa; Jose Luis Perez-Gracia; A Sánchez-Arráez; David Sancho; Ignacio Melero

Dendritic cells (DCs) are the main professional antigen-presenting cells for induction of T-cell adaptive responses. Cancer cells express tumor antigens, including neoantigens generated by nonsynonymous mutations, but are poor for antigen presentation and for providing costimulatory signals for T-cell priming. Mounting evidence suggests that antigen transfer to DCs and their surrogate presentation on major histocompatibility complex class I and II molecules together with costimulatory signals is paramount for induction of viral and cancer immunity. Of the great diversity of DCs, BATF3/IRF8-dependent conventional DCs type 1 (cDC1) excel at cross-presentation of tumor cell-associated antigens. Location of cDC1s in the tumor correlates with improved infiltration by CD8+ T cells and tumor-specific T-cell immunity. Indeed, cDC1s are crucial for antitumor efficacy using checkpoint inhibitors and anti-CD137 agonist monoclonal antibodies in mouse models. Enhancement and exploitation of T-cell cross-priming by cDC1s offer opportunities for improved cancer immunotherapy, including in vivo targeting of tumor antigens to internalizing receptors on cDC1s and strategies to increase their numbers, activation and priming capacity within tumors and tumor-draining lymph nodes.


Cancer immunology research | 2018

CD137 (4-1BB) costimulation modifies DNA methylation in CD8+ T-cell relevant genes.

M. Angela Aznar; Sara Labiano; Angel Diaz-Lagares; Carmen Molina; Saray Garasa; Arantza Azpilikueta; Iñaki Etxeberria; Alfonso R. Sánchez-Paulete; Alan J. Korman; Manel Esteller; Juan Sandoval; Ignacio Melero

CD137(4-1BB) ligation with agonist monoclonal antibodies used in immunotherapy induces long-term sequence-specific CpG DNA-methylation patterns in human CD8+ T lymphocytes. These epigenetic changes give rise to mRNA and protein expression changes in key T-cell genes and regulatory factors. CD137 (4-1BB) costimulation imprints long-term changes that instruct the ultimate behavior of T cells that have previously experienced CD137 ligation. Epigenetic changes could provide a suitable mechanism for these long-term consequences. Genome-wide DNA methylation arrays were carried out on human peripheral blood CD8+ T lymphocytes stimulated with agonist monoclonal antibody to CD137, including urelumab, which is in phase I/II clinical trials for cancer immunotherapy. Several genes showed consistent methylation patterns in response to CD137 costimulation, which were confirmed by pyrosequencing in a series of healthy donors. CD96, HHLA2, CCR5, CXCR5, and CCL5 were among the immune-related genes regulated by differential DNA methylation, leading to changes in mRNA and protein expression. These genes are also differentially methylated in naïve versus antigen-experienced CD8+ T cells. The transcription factor TCF1 and the microRNA miR-21 were regulated by DNA methylation upon CD137 costimulation. Such gene-expression regulatory factors can, in turn, broaden the effects of DNA methylation by controlling expression of their target genes. Overall, chromatin remodeling is postulated to leave CD137-costimulated T lymphocytes poised to differentially respond upon subsequent antigen recognition. Accordingly, CD137 connects costimulation during priming to genome-wide DNA methylation and chromatin reprogramming. Cancer Immunol Res; 6(1); 69–78. ©2017 AACR.

Collaboration


Dive into the Alfonso R. Sánchez-Paulete's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge