Network


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

Hotspot


Dive into the research topics where Isabella C. Glitza is active.

Publication


Featured researches published by Isabella C. Glitza.


Cancer Discovery | 2016

Loss of PTEN promotes resistance to T cell–mediated immunotherapy

Weiyi Peng; Jie Qing Chen; Chengwen Liu; Shruti Malu; Caitlin Creasy; Michael T. Tetzlaff; Chunyu Xu; Jodi A. McKenzie; Chunlei Zhang; Xiaoxuan Liang; Leila Williams; Wanleng Deng; Guo Chen; Rina M. Mbofung; Alexander J. Lazar; Carlos A. Torres-Cabala; Zachary A. Cooper; Pei-Ling Chen; Trang Tieu; Stefani Spranger; Xiaoxing Yu; Chantale Bernatchez; Marie-Andree Forget; Cara Haymaker; Rodabe N. Amaria; Jennifer L. McQuade; Isabella C. Glitza; Tina Cascone; Haiyan S. Li; Lawrence N. Kwong

UNLABELLED T cell-mediated immunotherapies are promising cancer treatments. However, most patients still fail to respond to these therapies. The molecular determinants of immune resistance are poorly understood. We show that loss of PTEN in tumor cells in preclinical models of melanoma inhibits T cell-mediated tumor killing and decreases T-cell trafficking into tumors. In patients, PTEN loss correlates with decreased T-cell infiltration at tumor sites, reduced likelihood of successful T-cell expansion from resected tumors, and inferior outcomes with PD-1 inhibitor therapy. PTEN loss in tumor cells increased the expression of immunosuppressive cytokines, resulting in decreased T-cell infiltration in tumors, and inhibited autophagy, which decreased T cell-mediated cell death. Treatment with a selective PI3Kβ inhibitor improved the efficacy of both anti-PD-1 and anti-CTLA-4 antibodies in murine models. Together, these findings demonstrate that PTEN loss promotes immune resistance and support the rationale to explore combinations of immunotherapies and PI3K-AKT pathway inhibitors. SIGNIFICANCE This study adds to the growing evidence that oncogenic pathways in tumors can promote resistance to the antitumor immune response. As PTEN loss and PI3K-AKT pathway activation occur in multiple tumor types, the results support the rationale to further evaluate combinatorial strategies targeting the PI3K-AKT pathway to increase the efficacy of immunotherapy.


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.


Science Translational Medicine | 2017

Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance

Whijae Roh; Pei Ling Chen; Alexandre Reuben; Christine N. Spencer; Peter A. Prieto; John P. Miller; Vancheswaran Gopalakrishnan; Feng Wang; Zachary A. Cooper; Sangeetha M. Reddy; Curtis Gumbs; Latasha Little; Qing Chang; Wei Shen Chen; Khalida Wani; Mariana Petaccia de Macedo; Eveline Chen; Jacob Austin-Breneman; Hong Jiang; Jason Roszik; Michael T. Tetzlaff; Michael A. Davies; Jeffrey E. Gershenwald; Hussein Abdul-Hassan Tawbi; Alexander J. Lazar; Patrick Hwu; Wen-Jen Hwu; Adi Diab; Isabella C. Glitza; Sapna Pradyuman Patel

Profiling of melanoma patients treated with checkpoint blockade reveals TCR clonality and copy number loss as correlates of therapeutic response. Checking on checkpoint inhibitors Immune checkpoint blockade has greatly improved the success of treatment in melanoma and other tumor types, but it is expensive and does not work for all patients. To optimize the likelihood of therapeutic success and reduce the risks and expense of unnecessary treatment, it would be helpful to find biomarkers that can predict treatment response. Roh et al. studied patients treated with sequential checkpoint inhibitors targeting CTLA-4 and then PD-1. In these patients, the authors discovered that a more clonal T cell population specifically correlates with response to PD-1 blockade, but not CTLA-4, which may help identify the best candidates for this treatment. In addition, increased frequency of gene copy number loss was correlated with decreased responsiveness to either therapy. Immune checkpoint blockade produces clinical benefit in many patients. However, better biomarkers of response are still needed, and mechanisms of resistance remain incompletely understood. To address this, we recently studied a cohort of melanoma patients treated with sequential checkpoint blockade against cytotoxic T lymphocyte antigen–4 (CTLA-4) followed by programmed death receptor–1 (PD-1) and identified immune markers of response and resistance. Building on these studies, we performed deep molecular profiling including T cell receptor sequencing and whole-exome sequencing within the same cohort and demonstrated that a more clonal T cell repertoire was predictive of response to PD-1 but not CTLA-4 blockade. Analysis of CNAs identified a higher burden of copy number loss in nonresponders to CTLA-4 and PD-1 blockade and found that it was associated with decreased expression of genes in immune-related pathways. The effect of mutational load and burden of copy number loss on response was nonredundant, suggesting the potential utility of a combinatorial biomarker to optimize patient care with checkpoint blockade therapy.


OncoImmunology | 2016

Distinct clinical patterns and immune infiltrates are observed at time of progression on targeted therapy versus immune checkpoint blockade for melanoma

Zachary A. Cooper; Alexandre Reuben; Christine N. Spencer; Peter A. Prieto; Jacob Austin-Breneman; Hong Jiang; Cara Haymaker; Vancheswaran Gopalakrishnan; Michael T. Tetzlaff; Dennie T. Frederick; Ryan J. Sullivan; Rodabe N. Amaria; Sapna Pradyuman Patel; Patrick Hwu; Scott E. Woodman; Isabella C. Glitza; Adi Diab; Luis Vence; Jaime Rodriguez-Canales; Edwin R. Parra; Ignacio I. Wistuba; Lisa M. Coussens; Arlene H. Sharpe; Keith T. Flaherty; Jeffrey E. Gershenwald; Lynda Chin; Michael A. Davies; Karen Clise-Dwyer; James P. Allison; Padmanee Sharma

ABSTRACT We have made major advances in the treatment of melanoma through the use of targeted therapy and immune checkpoint blockade; however, clinicians are posed with therapeutic dilemmas regarding timing and sequence of therapy. There is a growing appreciation of the impact of antitumor immune responses to these therapies, and we performed studies to test the hypothesis that clinical patterns and immune infiltrates differ at progression on these treatments. We observed rapid clinical progression kinetics in patients on targeted therapy compared to immune checkpoint blockade. To gain insight into possible immune mechanisms behind these differences, we performed deep immune profiling in tumors of patients on therapy. We demonstrated low CD8+ T-cell infiltrate on targeted therapy and high CD8+ T-cell infiltrate on immune checkpoint blockade at clinical progression. These data have important implications, and suggest that antitumor immune responses should be assessed when considering therapeutic options for patients with melanoma.


Lancet Oncology | 2018

Neoadjuvant plus adjuvant dabrafenib and trametinib versus standard of care in patients with high-risk, surgically resectable melanoma: a single-centre, open-label, randomised, phase 2 trial

Rodabe N. Amaria; Peter A. Prieto; Michael T. Tetzlaff; Alexandre Reuben; Miles C. Andrews; Merrick I. Ross; Isabella C. Glitza; Janice N. Cormier; Wen-Jen Hwu; Hussein Abdul-Hassan Tawbi; Sapna Pradyuman Patel; Jeffrey E. Lee; Jeffrey E. Gershenwald; Christine N. Spencer; Vancheswaran Gopalakrishnan; Roland L. Bassett; Lauren Simpson; Rosalind Mouton; Courtney W. Hudgens; Li Zhao; Haifeng Zhu; Zachary A. Cooper; Khalida Wani; Alexander J. Lazar; Patrick Hwu; Adi Diab; Michael K. Wong; Jennifer L. McQuade; Richard E. Royal; Anthony Lucci

BACKGROUND Dual BRAF and MEK inhibition produces a response in a large number of patients with stage IV BRAF-mutant melanoma. The existing standard of care for patients with clinical stage III melanoma is upfront surgery and consideration for adjuvant therapy, which is insufficient to cure most patients. Neoadjuvant targeted therapy with BRAF and MEK inhibitors (such as dabrafenib and trametinib) might provide clinical benefit in this high-risk p opulation. METHODS We undertook this single-centre, open-label, randomised phase 2 trial at the University of Texas MD Anderson Cancer Center (Houston, TX, USA). Eligible participants were adult patients (aged ≥18 years) with histologically or cytologically confirmed surgically resectable clinical stage III or oligometastatic stage IV BRAFV600E or BRAFV600K (ie, Val600Glu or Val600Lys)-mutated melanoma. Eligible patients had to have an Eastern Cooperative Oncology Group performance status of 0 or 1, a life expectancy of more than 3 years, and no previous exposure to BRAF or MEK inhibitors. Exclusion criteria included metastases to bone, brain, or other sites where complete surgical excision was in doubt. We randomly assigned patients (1:2) to either upfront surgery and consideration for adjuvant therapy (standard of care group) or neoadjuvant plus adjuvant dabrafenib and trametinib (8 weeks of neoadjuvant oral dabrafenib 150 mg twice per day and oral trametinib 2 mg per day followed by surgery, then up to 44 weeks of adjuvant dabrafenib plus trametinib starting 1 week after surgery for a total of 52 weeks of treatment). Randomisation was not masked and was implemented by the clinical trial conduct website maintained by the trial centre. Patients were stratified by disease stage. The primary endpoint was investigator-assessed event-free survival (ie, patients who were alive without disease progression) at 12 months in the intent-to-treat population. This trial is registered at ClinicalTrials.gov, number NCT02231775. FINDINGS Between Oct 23, 2014, and April 13, 2016, we randomly assigned seven patients to standard of care, and 14 to neoadjuvant plus adjuvant dabrafenib and trametinib. The trial was stopped early after a prespecified interim safety analysis that occurred after a quarter of the participants had been accrued revealed significantly longer event-free survival with neoadjuvant plus adjuvant dabrafenib and trametinib than with standard of care. After a median follow-up of 18·6 months (IQR 14·6-23·1), significantly more patients receiving neoadjuvant plus adjuvant dabrafenib and trametinib were alive without disease progression than those receiving standard of care (ten [71%] of 14 patients vs none of seven in the standard of care group; median event-free survival was 19·7 months [16·2-not estimable] vs 2·9 months [95% CI 1·7-not estimable]; hazard ratio 0·016, 95% CI 0·00012-0·14, p<0·0001). Neoadjuvant plus adjuvant dabrafenib and trametinib were well tolerated with no occurrence of grade 4 adverse events or treatment-related deaths. The most common adverse events in the neoadjuvant plus adjuvant dabrafenib and trametinib group were expected grade 1-2 toxicities including chills (12 patients [92%]), headache (12 [92%]), and pyrexia (ten [77%]). The most common grade 3 adverse event was diarrhoea (two patients [15%]). INTERPRETATION Neoadjuvant plus adjuvant dabrafenib and trametinib significantly improved event-free survival versus standard of care in patients with high-risk, surgically resectable, clinical stage III-IV melanoma. Although the trial finished early, limiting generalisability of the results, the findings provide proof-of-concept and support the rationale for further investigation of neoadjuvant approaches in this disease. This trial is currently continuing accrual as a single-arm study of neoadjuvant plus adjuvant dabrafenib and trametinib. FUNDING Novartis Pharmaceuticals Corporation.


Pigment Cell & Melanoma Research | 2016

Melanoma central nervous system metastases: current approaches, challenges, and opportunities

Justine V. Cohen; Hussain Tawbi; Kim Margolin; Ravi Amravadi; Marcus Bosenberg; Priscilla K. Brastianos; Veronica L. Chiang; John F. de Groot; Isabella C. Glitza; Meenhard Herlyn; Sheri L. Holmen; Lucia B. Jilaveanu; Andrew B. Lassman; Stergios J. Moschos; Michael A. Postow; Reena Thomas; John Tsiouris; Patrick Y. Wen; Richard M. White; Timothy Turnham; Michael A. Davies; Harriet M. Kluger

Melanoma central nervous system metastases are increasing, and the challenges presented by this patient population remain complex. In December 2015, the Melanoma Research Foundation and the Wistar Institute hosted the First Summit on Melanoma Central Nervous System (CNS) Metastases in Philadelphia, Pennsylvania. Here, we provide a review of the current status of the field of melanoma brain metastasis research; identify key challenges and opportunities for improving the outcomes in patients with melanoma brain metastases; and set a framework to optimize future research in this critical area.


Leukemia & Lymphoma | 2015

Chromosome 8q24.1/c-MYC abnormality: a marker for high-risk myeloma

Isabella C. Glitza; Gary Lu; Rupin Shah; Qaiser Bashir; Nina Shah; Richard E. Champlin; Jatin J. Shah; Robert Z. Orlowski; Muzaffar H. Qazilbash

Abstract The proto-oncogene c-MYC is rearranged in about 15% of patients with multiple myeloma (MM). We identified 23 patients with MM and c-MYC. Primary objectives were to describe the clinical characteristics, response to therapy, progression-free survival and overall survival (OS). Twelve out of twenty-three patients presented with or progressed to either plasma cell leukemia (PCL) and/or extramedullary disease (EMD). Induction therapy consisted of an immunomodulatory, proteasome inhibitor-based or conventional chemotherapy regimen. Fifteen patients achieved a partial response and three achieved a very good partial response. Sixteen patients received an autologous and one patient an allogeneic hematopoietic stem cell transplant. Median OS from diagnosis was 20.2 months. Patients with PCL or EMD had significantly shorter OS (15.5 vs. 40.4 months, p = 0.0005). This is the first report describing the clinical characteristics of patients with MM and c-MYC. These abnormalities are associated with an aggressive form of MM, high incidence of PCL/EMD and short OS.


npj Genomic Medicine | 2017

Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma

Alexandre Reuben; Christine N. Spencer; Peter A. Prieto; Vancheswaran Gopalakrishnan; Sangeetha M. Reddy; John P. Miller; Xizeng Mao; Mariana Petaccia de Macedo; Jiong Chen; Xingzhi Song; Hong Jiang; Pei Ling Chen; Hannah C. Beird; Haven R. Garber; Whijae Roh; Khalida Wani; Eveline Chen; Cara Haymaker; Marie Andrée Forget; Latasha Little; Curtis Gumbs; Rebecca Thornton; Courtney W. Hudgens; Wei Shen Chen; Jacob Austin-Breneman; Robert Sloane; Luigi Nezi; Alexandria P. Cogdill; Chantale Bernatchez; Jason Roszik

Appreciation for genomic and immune heterogeneity in cancer has grown though the relationship of these factors to treatment response has not been thoroughly elucidated. To better understand this, we studied a large cohort of melanoma patients treated with targeted therapy or immune checkpoint blockade (n = 60). Heterogeneity in therapeutic responses via radiologic assessment was observed in the majority of patients. Synchronous melanoma metastases were analyzed via deep genomic and immune profiling, and revealed substantial genomic and immune heterogeneity in all patients studied, with considerable diversity in T cell frequency, and few shared T cell clones (<8% on average) across the cohort. Variables related to treatment response were identified via these approaches and through novel radiomic assessment. These data yield insight into differential therapeutic responses to targeted therapy and immune checkpoint blockade in melanoma, and have key translational implications in the age of precision medicine.Melanoma: Tumor differences within a patient may explain heterogeneous responsesPatients with metastatic melanoma display molecular and immune differences across tumor sites associated with differential drug responses. A team led by Jennifer Wargo from the University of Texas MD Anderson Cancer Center, Houston, USA, studied the radiological responses of 60 patients with metastatic melanoma, half of whom received targeted drug therapy and half of whom received an immune checkpoint inhibitor. The majority (83%) showed differences in responses across metastases. The group then profiled tumors in a subset, and found molecular and immune heterogeneity in different tumors within the same patient. Heterogeneity in mutational and immune profiles within tumors from individual patients could explain differences in treatment response. Knowing this, the authors emphasize the importance of acquiring biopsies from more than one tumor site in order to best tailor therapies to the features of metastatic cancer.


Archives of Pathology & Laboratory Medicine | 2013

Plasma cell enrichment enhances detection of high-risk cytogenomic abnormalities by fluorescence in situ hybridization and improves risk stratification of patients with plasma cell neoplasms.

Gary Lu; Ramya Muddasani; Robert Z. Orlowski; Lynne V. Abruzzo; Muzaffar H. Qazilbash; M. James You; Yaping Wang; Ming Zhao; Su Chen; Isabella C. Glitza; L. Jeffrey Medeiros

CONTEXT Methods for plasma cell enrichment of bone marrow (BM) specimens can increase the sensitivity of fluorescence in situ hybridization (FISH) for detecting cytogenomic abnormalities. There are no published reports using these methods to evaluate high-risk cytogenomic abnormalities in patients with plasma cell neoplasms (PCNs) after therapy. OBJECTIVE To evaluate the utility of plasma cell enrichment combined with FISH for detection of high-risk cytogenomic abnormalities in patients with PCNs after therapy. DESIGN Twenty-eight patients with PCNs, of whom 22 received treatment, were included in this study. Plasma cells were enriched in BM aspirates by using a magnetic cell-sorting procedure to select CD138(+) cells. Probes were chosen to assess for del(17p13/TP53), del(13q14/RB1), 1q21/CKS1B gain, IgH/FGFR3, and IgH/MAF. Clinicopathologic data were collected during clinical follow-up after plasma cell enrichment. RESULTS Plasma cells in nonenriched BM specimens ranged from 1% to 28% (median, 8%) compared with 28% to 96% (median, 73%) in enriched BM specimens (P < .001). In a subset of treated patients in clinical remission, FISH detected high-risk cytogenomic abnormalities only in plasma cell-enriched samples. This approach also detected abnormalities in cases of solitary plasmacytoma and monoclonal gammopathy of undetermined significance. CONCLUSIONS Plasma cell enrichment of BM specimens increases FISH sensitivity for detecting high-risk cytogenomic abnormalities, particularly in treated patients, and these results, in combination with clinical follow-up data, can be of value to improve risk stratification and patient management.

Collaboration


Dive into the Isabella C. Glitza's collaboration.

Top Co-Authors

Avatar

Sapna Pradyuman Patel

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Patrick Hwu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rodabe N. Amaria

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

Adi Diab

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael A. Davies

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hussein Abdul-Hassan Tawbi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Scott E. Woodman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael T. Tetzlaff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer A. Wargo

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge