Network


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

Hotspot


Dive into the research topics where Luis Vence is active.

Publication


Featured researches published by Luis Vence.


Lancet Oncology | 2014

Safety and Activity of PD1 Blockade by Pidilizumab in Combination with Rituximab in Patients with Relapsed Follicular Lymphoma: a Single Group, Open-label, Phase 2 Trial

Jason R. Westin; Fuliang Chu; Min Zhang; Luis Fayad; Larry W. Kwak; Nathan Fowler; Jorge Romaguera; Fredrick B. Hagemeister; Michelle A. Fanale; Felipe Samaniego; Lei Feng; Veerabhadran Baladandayuthapani; Zhiqiang Wang; Wencai Ma; Yanli Gao; Michael J. Wallace; Luis Vence; Laszlo Radvanyi; Tariq Muzzafar; Rinat Rotem-Yehudar; R. Eric Davis; Sattva S. Neelapu

BACKGROUND Endogenous or iatrogenic antitumour immune responses can improve the course of follicular lymphoma, but might be diminished by immune checkpoints in the tumour microenvironment. These checkpoints might include effects of programmed cell death 1 (PD1), a co-inhibitory receptor that impairs T-cell function and is highly expressed on intratumoral T cells. We did this phase 2 trial to investigate the activity of pidilizumab, a humanised anti-PD1 monoclonal antibody, with rituximab in patients with relapsed follicular lymphoma. METHODS We did this open-label, non-randomised trial at the University of Texas MD Anderson Cancer Center (Houston, TX, USA). Adult (≥18 years) patients with rituximab-sensitive follicular lymphoma relapsing after one to four previous therapies were eligible. Pidilizumab was administered at 3 mg/kg intravenously every 4 weeks for four infusions, plus eight optional infusions every 4 weeks for patients with stable disease or better. Starting 17 days after the first infusion of pidilizumab, rituximab was given at 375 mg/m(2) intravenously weekly for 4 weeks. The primary endpoint was the proportion of patients who achieved an objective response (complete response plus partial response according to Revised Response Criteria for Malignant Lymphoma). Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00904722. FINDINGS We enrolled 32 patients between Jan 13, 2010, and Jan 20, 2012. Median follow-up was 15.4 months (IQR 10.1-21.0). The combination of pidilizumab and rituximab was well tolerated, with no autoimmune or treatment-related adverse events of grade 3 or 4. The most common adverse events of grade 1 were anaemia (14 patients) and fatigue (13 patients), and the most common adverse event of grade 2 was respiratory infection (five patients). Of the 29 patients evaluable for activity, 19 (66%) achieved an objective response: complete responses were noted in 15 (52%) patients and partial responses in four (14%). INTERPRETATION The combination of pidilizumab plus rituximab is well tolerated and active in patients with relapsed follicular lymphoma. Our results suggest that immune checkpoint blockade is worthy of further study in follicular lymphoma. FUNDING National Institutes of Health, Leukemia and Lymphoma Society, Cure Tech, and University of Texas MD Anderson Cancer Center.


Science | 2018

Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients.

V. Gopalakrishnan; C. N. Spencer; Luigi Nezi; Alexandre Reuben; Miles C. Andrews; T. V. Karpinets; Peter A. Prieto; D. Vicente; K. Hoffman; Spencer C. Wei; Alexandria P. Cogdill; Li Zhao; Courtney W. Hudgens; D. S. Hutchinson; T. Manzo; M. Petaccia de Macedo; Tiziana Cotechini; T. Kumar; Wei Shen Chen; Sangeetha M. Reddy; R. Szczepaniak Sloane; J. Galloway-Pena; Hong Jiang; Pei Ling Chen; E. J. Shpall; K. Rezvani; A. M. Alousi; R. F. Chemaly; S. Shelburne; Luis Vence

Good bacteria help fight cancer Resident gut bacteria can affect patient responses to cancer immunotherapy (see the Perspective by Jobin). Routy et al. show that antibiotic consumption is associated with poor response to immunotherapeutic PD-1 blockade. They profiled samples from patients with lung and kidney cancers and found that nonresponding patients had low levels of the bacterium Akkermansia muciniphila. Oral supplementation of the bacteria to antibiotic-treated mice restored the response to immunotherapy. Matson et al. and Gopalakrishnan et al. studied melanoma patients receiving PD-1 blockade and found a greater abundance of “good” bacteria in the guts of responding patients. Nonresponders had an imbalance in gut flora composition, which correlated with impaired immune cell activity. Thus, maintaining healthy gut flora could help patients combat cancer. Science, this issue p. 91, p. 104, p. 97; see also p. 32 Gut bacteria influence patient response to cancer therapy. Preclinical mouse models suggest that the gut microbiome modulates tumor response to checkpoint blockade immunotherapy; however, this has not been well-characterized in human cancer patients. Here we examined the oral and gut microbiome of melanoma patients undergoing anti–programmed cell death 1 protein (PD-1) immunotherapy (n = 112). Significant differences were observed in the diversity and composition of the patient gut microbiome of responders versus nonresponders. Analysis of patient fecal microbiome samples (n = 43, 30 responders, 13 nonresponders) showed significantly higher alpha diversity (P < 0.01) and relative abundance of bacteria of the Ruminococcaceae family (P < 0.01) in responding patients. Metagenomic studies revealed functional differences in gut bacteria in responders, including enrichment of anabolic pathways. Immune profiling suggested enhanced systemic and antitumor immunity in responding patients with a favorable gut microbiome as well as in germ-free mice receiving fecal transplants from responding patients. Together, these data have important implications for the treatment of melanoma patients with immune checkpoint inhibitors.


Cancer Discovery | 2016

Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade

Pei Ling Chen; Whijae Roh; Alexandre Reuben; Zachary A. Cooper; Christine N. Spencer; Peter A. Prieto; John P. Miller; Roland L. Bassett; Vancheswaran Gopalakrishnan; Khalida Wani; Mariana Petaccia de Macedo; Jacob Austin-Breneman; Hong Jiang; Qing Chang; Sangeetha M. Reddy; Wei Shen Chen; Michael T. Tetzlaff; R. Broaddus; Michael A. Davies; Jeffrey E. Gershenwald; Lauren E. Haydu; Alexander J. Lazar; Sapna Pradyuman Patel; Patrick Hwu; Wen-Jen Hwu; Adi Diab; Isabella C. Glitza; Scott E. Woodman; Luis Vence; Ignacio I. Wistuba

UNLABELLED Immune checkpoint blockade represents a major breakthrough in cancer therapy; however, responses are not universal. Genomic and immune features in pretreatment tumor biopsies have been reported to correlate with response in patients with melanoma and other cancers, but robust biomarkers have not been identified. We studied a cohort of patients with metastatic melanoma initially treated with cytotoxic T-lymphocyte-associated antigen-4 (CTLA4) blockade (n = 53) followed by programmed death-1 (PD-1) blockade at progression (n = 46), and analyzed immune signatures in longitudinal tissue samples collected at multiple time points during therapy. In this study, we demonstrate that adaptive immune signatures in tumor biopsy samples obtained early during the course of treatment are highly predictive of response to immune checkpoint blockade and also demonstrate differential effects on the tumor microenvironment induced by CTLA4 and PD-1 blockade. Importantly, potential mechanisms of therapeutic resistance to immune checkpoint blockade were also identified. SIGNIFICANCE These studies demonstrate that adaptive immune signatures in early on-treatment tumor biopsies are predictive of response to checkpoint blockade and yield insight into mechanisms of therapeutic resistance. These concepts have far-reaching implications in this age of precision medicine and should be explored in immune checkpoint blockade treatment across cancer types. Cancer Discov; 6(8); 827-37. ©2016 AACR.See related commentary by Teng et al., p. 818This article is highlighted in the In This Issue feature, p. 803.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Circulating tumor antigen-specific regulatory T cells in patients with metastatic melanoma

Luis Vence; A. Karolina Palucka; Joseph W. Fay; Yong-Jun Liu; Jacques Banchereau; Hideki Ueno

Although it is accepted that regulatory T cells (T regs) contribute to cancer progression, most studies in the field consider nonantigen-specific suppression. Here, we show the presence of tumor antigen-specific CD4+ T regs in the blood of patients with metastatic melanoma. These CD4+ T regs recognize a broad range of tumor antigens, including gp100 and TRP1 (melanoma tissue differentiation antigens), NY-ESO-1 (cancer/testis antigen) and survivin (inhibitor of apoptosis protein (IAP) family antigen). These tumor antigen-specific T regs proliferate in peripheral blood mononuclear cells (PBMC) cultures in response to specific 15-mer peptides, produce preferentially IL-10 and express high levels of FoxP3. They suppress autologous CD4+CD25− T cell responses in a cell contact-dependent manner and thus share properties of both naturally occurring regulatory T cells and type 1 regulatory T cells. Such tumor antigen-specific T regs were not detected in healthy individuals. These tumor antigen-specific T regs might thus represent another target for immunotherapy of metastatic melanoma.


Nature Communications | 2014

The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis

Roberto Lande; Elisabetta Botti; Camilla Jandus; Danijel Dojcinovic; Giorgia Fanelli; Curdin Conrad; Georgios Chamilos; Laurence Feldmeyer; Barbara Marinari; Susan Chon; Luis Vence; Valeria Riccieri; Phillippe Guillaume; Alex A. Navarini; Pedro Romero; Antonio Costanzo; Enza Piccolella; Michel Gilliet; Loredana Frasca

Psoriasis is a common T-cell-mediated skin disease with 2-3% prevalence worldwide. Psoriasis is considered to be an autoimmune disease, but the precise nature of the autoantigens triggering T-cell activation remains poorly understood. Here we find that two-thirds of patients with moderate-to-severe plaque psoriasis harbour CD4(+) and/or CD8(+) T cells specific for LL37, an antimicrobial peptide (AMP) overexpressed in psoriatic skin and reported to trigger activation of innate immune cells. LL37-specific T cells produce IFN-γ, and CD4(+) T cells also produce Th17 cytokines. LL37-specific T cells can infiltrate lesional skin and may be tracked in patients blood by tetramers staining. Presence of circulating LL37-specific T cells correlates significantly with disease activity, suggesting a contribution to disease pathogenesis. Thus, we uncover a role of LL37 as a T-cell autoantigen in psoriasis and provide evidence for a role of AMPs in both innate and adaptive immune cell activation.


Clinical Cancer Research | 2012

BRAF(V600) Inhibitor GSK2118436 Targeted Inhibition of Mutant BRAF in Cancer Patients Does Not Impair Overall Immune Competency

David S. Hong; Luis Vence; Gerald S. Falchook; Laszlo Radvanyi; Chengwen Liu; Vicki L. Goodman; Jeffery J. Legos; Sam Blackman; Antonio Scarmadio; Razelle Kurzrock; Gregory Lizée; Patrick Hwu

Purpose: An intact immune system likely contributes to the outcome of treatment and may be important for clearance of drug-resistant tumor cells and for prevention of recurrence. Although pharmacologic inhibition of BRAF(V600E) in melanoma patients, which is linked to immune suppression, results in an initial response rate, these responses are typically of limited duration. Combining immunotherapeutic drugs with kinase-targeted agents is an attractive strategy to increase clinical efficacy. Evidence suggesting that mitogen-activated protein kinase pathway activation in tumor cells contributes to immune suppression suggests that the two approaches may be synergistic, provided that BRAF(V600E) inhibitors are nontoxic to immune cells. Methods: To assess effects of mutant BRAF inhibition on systemic immunity, we studied 13 patients with tumors carrying a BRAF mutation who underwent treatment with GSK2118436, a V600 mutant BRAF-specific inhibitor. We carried out peripheral blood immunomonitoring before and following one or two 28-day cycles of treatment. Results: GSK2118436 treatment had no detectable impact on most immune parameters tested, including serum cytokine levels, peripheral blood cell counts, leukocyte subset frequencies, and memory CD4+ and CD8+ T-cell recall responses. A slight increase in serum TNF-α over the course of treatment was observed. In addition, three of the four human leukocyte antigen-A2–positive patients experienced a modest increase in circulating tumor antigen–specific CD8+ T cells following BRAF(V600) inhibitor therapy. Conclusions: GSK2118436 treatment results in no detectable negative impact on existing systemic immunity or the de novo generation of tumor-specific T cells. These findings suggest that future trials combining specific BRAF(V600E) inhibition with immunotherapy should not impair immune response. Clin Cancer Res; 18(8); 2326–35. ©2012 AACR.


Journal of Clinical Investigation | 2014

IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients

Geok Choo Sim; Lei Jin; Yan Yang; Sheng Wu; Edwina Washington; Deborah Sanders; Carol Lacey; Yijun Wang; Luis Vence; Patrick Hwu; Laszlo Radvanyi

High-dose (HD) IL-2 therapy in patients with cancer increases the general population of Tregs, which are positive for CD4, CD25, and the Treg-specific marker Foxp3. It is unknown whether specific subsets of Tregs are activated and expanded during HD IL-2 therapy or whether activation of any particular Treg subset correlates with clinical outcome. Here, we evaluated Treg population subsets that were induced in patients with melanoma following HD IL-2 therapy. We identified a Treg population that was positive for CD4, CD25, Foxp3, and the inducible T cell costimulator (ICOS). This Treg population increased more than any other lymphocyte subset during HD IL-2 therapy and had an activated Treg phenotype, as indicated by high levels of CD39, CD73, and TGF-β. ICOS(+) Tregs were the most proliferative lymphocyte population in the blood after IL-2 therapy. Patients with melanoma with enhanced expansion of ICOS(+) Tregs in blood following the first cycle of HD IL-2 therapy had worse clinical outcomes than patients with fewer ICOS(+) Tregs. However, there was no difference in total Treg expansion between HD IL-2 responders and nonresponders. These data suggest that increased expansion of the ICOS(+) Treg population following the first cycle of HD IL-2 therapy may be predictive of clinical outcome.


PLOS ONE | 2012

Depressive Symptoms and Cortisol Rhythmicity Predict Survival in Patients with Renal Cell Carcinoma: Role of Inflammatory Signaling

Lorenzo Cohen; Steven W. Cole; Anil K. Sood; Sarah Prinsloo; Clemens Kirschbaum; Jesusa M.G. Arevalo; Nicholas B. Jennings; Shellie M. Scott; Luis Vence; Qi Wei; Diane Kentor; Laszlo Radvanyi; Nizar M. Tannir; Eric Jonasch; Pheroze Tamboli; Louis L. Pisters

Purpose Evidence has supported the association between psychological factors and cancer biology; however, findings are equivocal on the role of psychosocial factors in cancer progression. This study generates a hypothesis of mechanistic variables by examining the clinical effects of psychosocial factors and cortisol dysregulation in patients with metastatic renal cell carcinoma (RCC) and examines associated activation of transcription control pathways. Methods Patients with metastatic RCC (n = 217) were prospectively enrolled in this study. Patients completed questionnaires (Centers for Epidemiologic Studies – Depression; SF-36 Health Status Survey; Duke Social Support Index; Coping Operations Preference Enquiry; organized and non-organized religious activity; and intrinsic religiosity), and provided blood and saliva samples. Cortisol levels and whole genome transcriptional profiling were assessed to identify potential alterations in circadian rhythms and genomic pathways. Results Separate Cox regression models, controlling for disease risk category, revealed that CES-D scores (p = 0.05, HR = 1.5, 95% CI for HR: 1.00–2.23) and cortisol slope (p = 0.002; HR = 1.9; 95%CI for HR: 1.27–2.97) were significantly associated with decreased survival. Only cortisol slope and risk category remained significant in the complete model. Functional genomic analyses linked depressive symptoms to increased expression of pro-inflammatory and pro-metastatic genes in circulating leukocytes. 116 transcripts were found to be upregulated by an average of 50% or more in high CES-D patients, and 57 transcripts downregulated by at least 50%. These changes were also found in the tumor in a subset of patients. Conclusion These findings identify depressive symptoms as a key predictor of survival in renal cell carcinoma patients with potential links to dysregulation of cortisol and inflammatory biology.


Nature Medicine | 2017

VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer

Jianjun Gao; John F. Ward; Curtis A. Pettaway; Lewis Zhichang Shi; Sumit K. Subudhi; Luis Vence; Hao Zhao; Jianfeng Chen; Hong Chen; Patricia Troncoso; James P. Allison; Christopher J. Logothetis; Ignacio I. Wistuba; Manuel A. Sepulveda; Jingjing Sun; Jennifer A. Wargo; Jorge Blando; Padmanee Sharma

To date, anti-CTLA-4 (ipilimumab) or anti-PD-1 (nivolumab) monotherapy has not been demonstrated to be of substantial clinical benefit in patients with prostate cancer. To identify additional immune-inhibitory pathways in the prostate-tumor microenvironment, we evaluated untreated and ipilimumab-treated tumors from patients in a presurgical clinical trial. Levels of the PD-L1 and VISTA inhibitory molecules increased on independent subsets of macrophages in treated tumors. Our data suggest that VISTA represents another compensatory inhibitory pathway in prostate tumors after ipilimumab therapy.


Cancer | 2011

Biologic and Immunologic Effects of Preoperative Trastuzumab for Ductal Carcinoma in Situ of the Breast

Henry M. Kuerer; Aman U. Buzdar; Elizabeth A. Mittendorf; Francisco J. Esteva; Anthony Lucci; Luis Vence; Laszlo Radvanyi; Funda Meric-Bernstam; Kelly K. Hunt; W. F. Symmans

Through this study, the authors sought to investigate the biologic and immunologic effects of preoperative trastuzumab in patients with ductal carcinoma in situ (DCIS) of the breast.

Collaboration


Dive into the Luis Vence's collaboration.

Top Co-Authors

Avatar

Padmanee Sharma

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

James P. Allison

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Hwu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jianjun Gao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge Blando

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chantale Bernatchez

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wen-Jen Hwu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cara Haymaker

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sapna Pradyuman Patel

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge