Network


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

Hotspot


Dive into the research topics where Michael D. Iglesia is active.

Publication


Featured researches published by Michael D. Iglesia.


Journal of Clinical Oncology | 2016

Molecular Heterogeneity and Response to Neoadjuvant Human Epidermal Growth Factor Receptor 2 Targeting in CALGB 40601, a Randomized Phase III Trial of Paclitaxel Plus Trastuzumab With or Without Lapatinib

Lisa A. Carey; Donald A. Berry; Constance Cirrincione; William T. Barry; Brandelyn N. Pitcher; Lyndsay Harris; David W. Ollila; Ian E. Krop; Norah Lynn Henry; Douglas Weckstein; Carey K. Anders; Baljit Singh; Katherine A. Hoadley; Michael D. Iglesia; Maggie Chon U. Cheang; Charles M. Perou; Clifford A. Hudis

PURPOSE Dual human epidermal growth factor receptor 2 (HER2) targeting can increase pathologic complete response rates (pCRs) to neoadjuvant therapy and improve progression-free survival in metastatic disease. CALGB 40601 examined the impact of dual HER2 blockade consisting of trastuzumab and lapatinib added to paclitaxel, considering tumor and microenvironment molecular features. PATIENTS AND METHODS Patients with stage II to III HER2-positive breast cancer underwent tumor biopsy followed by random assignment to paclitaxel plus trastuzumab alone (TH) or with the addition of lapatinib (THL) for 16 weeks before surgery. An investigational arm of paclitaxel plus lapatinib (TL) was closed early. The primary end point was pCR in the breast; correlative end points focused on molecular features identified by gene expression-based assays. RESULTS Among 305 randomly assigned patients (THL, n = 118; TH, n = 120; TL, n = 67), the pCR rate was 56% (95% CI, 47% to 65%) with THL and 46% (95% CI, 37% to 55%) with TH (P = .13), with no effect of dual therapy in the hormone receptor-positive subset but a significant increase in pCR with dual therapy in those with hormone receptor-negative disease (P = .01). The tumors were molecularly heterogeneous by gene expression analysis using mRNA sequencing (mRNAseq). pCR rates significantly differed by intrinsic subtype (HER2 enriched, 70%; luminal A, 34%; luminal B, 36%; P < .001). In multivariable analysis treatment arm, intrinsic subtype, HER2 amplicon gene expression, p53 mutation signature, and immune cell signatures were independently associated with pCR. Post-treatment residual disease was largely luminal A (69%). CONCLUSION pCR to dual HER2-targeted therapy was not significantly higher than single HER2 targeting. Tissue analysis demonstrated a high degree of intertumoral heterogeneity with respect to both tumor genomics and tumor microenvironment that significantly affected pCR rates. These factors should be considered when interpreting and designing trials in HER2-positive disease.


Clinical Cancer Research | 2014

Prognostic B-Cell Signatures using mRNA-Seq in Patients with Subtype-Specific Breast and Ovarian Cancer

Michael D. Iglesia; Benjamin G. Vincent; Joel S. Parker; Katherine A. Hoadley; Lisa A. Carey; Charles M. Perou; Jonathan S. Serody

Purpose: Lymphocytic infiltration of tumors predicts improved survival in patients with breast cancer. Previous studies have suggested that this survival benefit is confined predominantly to the basal-like subtype. Immune infiltration in ovarian tumors is also associated with improved prognosis. Currently, it is unclear what aspects of the immune response mediate this improved outcome. Experimental Design: Using The Cancer Genome Atlas mRNA-seq data and a large microarray dataset, we evaluated adaptive immune gene expression by genomic subtype in breast and ovarian cancer. To investigate B-cells observed to be prognostic within specific subtypes, we developed methods to analyze B-cell population diversity and degree of somatic hypermutation (SHM) from B-cell receptor (BCR) sequences in mRNA-seq data. Results: Improved metastasis-free/progression-free survival was correlated with B-cell gene expression signatures, which were restricted mainly to the basal-like and HER2-enriched breast cancer subtypes and the immunoreactive ovarian cancer subtype. Consistent with a restricted epitope-driven response, a subset of basal-like and HER2-enriched breast tumors and immunoreactive ovarian tumors showed high expression of a low-diversity population of BCR gene segments. More BCR segments showed improved prognosis with increased expression in basal-like breast tumors and immunoreactive ovarian tumors compared with other subtypes. Basal-like and HER2-enriched tumors exhibited more BCR sequence variants in regions consistent with SHM. Conclusion: Taken together, these data suggest the presence of a productive and potentially restricted antitumor B-cell response in basal-like breast and immunoreactive ovarian cancers. Immunomodulatory therapies that support B-cell responses may be a promising therapeutic approach to targeting these B-cell infiltrated tumors. Clin Cancer Res; 20(14); 3818–29. ©2014 AACR.


Journal of the National Cancer Institute | 2016

Genomic Analysis of Immune Cell Infiltrates Across 11 Tumor Types

Michael D. Iglesia; Joel S. Parker; Katherine A. Hoadley; Jonathan S. Serody; Charles M. Perou; Benjamin G. Vincent

BACKGROUND Immune infiltration of the tumor microenvironment has been associated with improved survival for some patients with solid tumors. The precise makeup and prognostic relevance of immune infiltrates across a broad spectrum of tumors remain unclear. METHODS Using mRNA sequencing data from The Cancer Genome Atlas (TCGA) from 11 tumor types representing 3485 tumors, we evaluated lymphocyte and macrophage gene expression by tissue type and by genomic subtypes defined within and across tumor tissue of origin (Cox proportional hazards, Pearson correlation). We investigated clonal diversity of B-cell infiltrates through calculating B-cell receptor (BCR) repertoire sequence diversity. All statistical tests were two-sided. RESULTS High expression of T-cell and B-cell signatures predicted improved overall survival across many tumor types including breast, lung, and melanoma (breast CD8_T_Cells hazard ratio [HR] = 0.36, 95% confidence interval [CI] = 0.16 to 0.81, P = .01; lung adenocarcinoma B_Cell_60gene HR = 0.71, 95% CI = 0.58 to 0.87, P = 7.80E-04; melanoma LCK HR = 0.86, 95% CI = 0.79 to 0.94, P = 6.75E-04). Macrophage signatures predicted worse survival in GBM, as did B-cell signatures in renal tumors (Glioblastoma Multiforme [GBM]: macrophages HR = 1.62, 95% CI = 1.17 to 2.26, P = .004; renal: B_Cell_60gene HR = 1.17, 95% CI = 1.04 to 1.32, P = .009). BCR diversity was associated with survival beyond gene segment expression in melanoma (HR = 2.67, 95% CI = 1.32 to 5.40, P = .02) and renal cell carcinoma (HR = 0.36, 95% CI = 0.15 to 0.87, P = .006). CONCLUSIONS These data support existing studies suggesting that in diverse tissue types, heterogeneous immune infiltrates are present and typically portend an improved prognosis. In some tumor types, BCR diversity was also associated with survival. Quantitative genomic signatures of immune cells warrant further testing as prognostic markers and potential biomarkers of response to cancer immunotherapy.


JCI insight | 2016

Claudin-low bladder tumors are immune infiltrated and actively immune suppressed

Jordan Kardos; Shengjie Chai; Lisle E. Mose; Sara R. Selitsky; Bhavani Krishnan; Ryoichi Saito; Michael D. Iglesia; Matthew I. Milowsky; Joel S. Parker; William Y. Kim; Benjamin G. Vincent

We report the discovery of a claudin-low molecular subtype of high-grade bladder cancer that shares characteristics with the homonymous subtype of breast cancer. Claudin-low bladder tumors were enriched for multiple genetic features including increased rates of RB1, EP300, and NCOR1 mutations; increased frequency of EGFR amplification; decreased rates of FGFR3, ELF3, and KDM6A mutations; and decreased frequency of PPARG amplification. While claudin-low tumors showed the highest expression of immune gene signatures, they also demonstrated gene expression patterns consistent with those observed in active immunosuppression. This did not appear to be due to differences in predicted neoantigen burden, but rather was associated with broad upregulation of cytokine and chemokine levels from low PPARG activity, allowing unopposed NFKB activity. Taken together, these results define a molecular subtype of bladder cancer with distinct molecular features and an immunologic profile that would, in theory, be primed for immunotherapeutic response.


Journal of Clinical Investigation | 2017

Treg depletion potentiates checkpoint inhibition in claudin-low breast cancer

Nicholas A. Taylor; Sarah C. Vick; Michael D. Iglesia; W. June Brickey; Bentley R. Midkiff; Karen P. McKinnon; Shannon Reisdorf; Carey K. Anders; Lisa A. Carey; Joel S. Parker; Charles M. Perou; Benjamin G. Vincent; Jonathan S. Serody

Claudin-low breast cancer is an aggressive subtype that confers poor prognosis and is found largely within the clinical triple-negative group of breast cancer patients. Here, we have shown that intrinsic and immune cell gene signatures distinguish the claudin-low subtype clinically as well as in mouse models of other breast cancer subtypes. Despite adaptive immune cell infiltration in claudin-low tumors, treatment with immune checkpoint inhibitory antibodies against cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death receptor 1 (PD-1) were ineffective in controlling tumor growth. CD4+FoxP3+ Tregs represented a large proportion of the tumor-infiltrating lymphocytes (TILs) in claudin-low tumors, and Tregs isolated from tumor-bearing mice were able to suppress effector T cell responses. Tregs in the tumor microenvironment highly expressed PD-1 and were recruited partly through tumor generation of the chemokine CXCL12. Antitumor efficacy required stringent Treg depletion combined with checkpoint inhibition; delays in tumor growth were not observed using therapies that modestly diminished the number of Tregs in the tumor microenvironment. This study provides evidence that the recruitment of Tregs to the tumor microenvironment inhibits an effective antitumor immune response and highlights early Treg recruitment as a possible mechanism for the lack of response to immune checkpoint blockade antibodies in specific subtypes of cancer that are heavily infiltrated with adaptive immune cells.


Bioinformatics | 2016

Assembly-based inference of B-cell receptor repertoires from short read RNA sequencing data with V'DJer.

Lisle E. Mose; Sara R. Selitsky; Lisa M. Bixby; David Marron; Michael D. Iglesia; Jonathan S. Serody; Charles M. Perou; Benjamin G. Vincent; Joel S. Parker

Motivation: B-cell receptor (BCR) repertoire profiling is an important tool for understanding the biology of diverse immunologic processes. Current methods for analyzing adaptive immune receptor repertoires depend upon PCR amplification of VDJ rearrangements followed by long read amplicon sequencing spanning the VDJ junctions. While this approach has proven to be effective, it is frequently not feasible due to cost or limited sample material. Additionally, there are many existing datasets where short-read RNA sequencing data are available but PCR amplified BCR data are not. Results: We present here V’DJer, an assembly-based method that reconstructs adaptive immune receptor repertoires from short-read RNA sequencing data. This method captures expressed BCR loci from a standard RNA-seq assay. We applied this method to 473 Melanoma samples from The Cancer Genome Atlas and demonstrate V’DJer’s ability to accurately reconstruct BCR repertoires from short read mRNA-seq data. Availability and Implementation: V’DJer is implemented in C/C ++, freely available for academic use and can be downloaded from Github: https://github.com/mozack/vdjer Contact: [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


Cancer Research | 2017

Abstract 1033: Estrogen receptor gene fusions drive endocrine therapy resistance in estrogen receptor positive breast cancer

Jonathan T. Lei; Jieya Shao; Jin Zhang; Michael D. Iglesia; Doug W. Chan; Ryoichi Matsunuma; Xiaping He; Purba Singh; Yoshimasa Kosaka; Robert Crowder; Svasti Haricharan; Shyam M. Kavuri; Jeremy Hoog; Chanpheng Phommaly; Rodrigo Franco Gonçalves; Susana Romalho; Wei-Chu Lai; Oliver A. Hampton; Anna Rogers; Ethan Tobias; Poojan Parikh; Sherri R. Davies; Cynthia X. Ma; Vera J. Suman; Kelly K. Hunt; Mark A. Watson; Katherine A. Hoadley; Aubrey E. Thompson; Charles M. Perou; Chad J. Creighton

Dysregulation of estrogen receptor gene (ESR1) is an established mechanism of inducing endocrine therapy resistance. We previously discovered a chromosomal translocation event generating an estrogen receptor gene fused in-frame to C-terminal sequences of YAP1 (ESR1-YAP1) that contributed to endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer models. This current study compares functional and pharmacological properties of additional ESR1 gene fusion events of both early stage (ESR1-NOP2) and advanced endocrine therapy resistant (ESR1-YAP1 and ESR1-PCDH11x) breast cancers. The YAP1 and PCDH11x fusions conferred estrogen-independent and fulvestrant-resistant growth in T47D, an ER+ breast cancer cell line in vitro and in vivo, in contrast to the NOP2 fusion which was sensitive to hormone deprivation. Immunohistochemical (IHC) staining of mouse lungs revealed significantly higher numbers of micrometastatic ER+ cells from the T47D tumors expressing the YAP1 and PCDH11x fusions than YFP control and NOP2 fusion. Estrogen response element (ERE) reporter and pull down assays revealed that although all ESR1 fusions studied bound EREs, only the YAP1 and PCDH11x caused ERE activation. Cell lines containing these “canonical” ESR1 fusions upregulated expression of ER responsive genes such as TFF1 and GREB1 in hormone deprived conditions. In contrast, the NOP2 fusion neither induced ERE activity nor upregulated TFF1 and GREB1 gene expression. The proliferative ability of canonical fusion-containing T47D cells was inhibited by palbociclib, a CDK4/6 inhibitor, in a dose-dependent manner. In vivo growth of patient-derived xenograft tumors naturally harboring the ESR1-YAP1 fusion (WHIM18) was significantly reduced in mice fed palbociclib-containing chow. Mice transplanted with WHIM18 also formed lung micrometastases, with an ER IHC staining pattern similar to lungs from YAP1 and PCDH11x fusion expressing T47D xenografts. In conclusion, in-frame ERE activating canonical fusions occur in end-stage, drug resistant, advanced breast cancer and can be added to ESR1 point mutations as a class of somatic mutation that may cause acquired resistance. Endocrine therapy resistant growth induced by these fusions can be treated with CDK4/6 inhibition, using an FDA approved drug, palbociclib, which could potentially improve outcomes in patients with ESR1 translocated tumors. Citation Format: Jonathan T. Lei, Jieya Shao, Jin Zhang, Michael Iglesia, Doug W. Chan, Ryoichi Matsunuma, Xiaping He, Purba Singh, Yoshimasa Kosaka, Robert Crowder, Svasti Haricharan, Shyam Kavuri, Jeremy Hoog, Chanpheng Phommaly, Rodrigo Goncalves, Susana Romalho, Wei-Chu Lai, Oliver Hampton, Anna Rogers, Ethan Tobias, Poojan Parikh, Sherri Davies, Cynthia Ma, Vera Suman, Kelly Hunt, Mark Watson, Katherine A. Hoadley, Aubrey Thompson, Charles Perou, Chad J. Creighton, Chris Maher, Matthew J. Ellis. Estrogen receptor gene fusions drive endocrine therapy resistance in estrogen receptor positive breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1033. doi:10.1158/1538-7445.AM2017-1033


Cancer Research | 2016

Abstract 5120: The novel claudin-low molecular subtype of high-grade urothelial bladder cancer is highly immunogenic yet immunosuppressed

Benjamin G. Vincent; William Y. Kim; Jordan Kardos; Shengjie Chai; Joel S. Parker; Lisle E. Mose; Sara R. Selitsky; Michael D. Iglesia; Matthew I. Milowsky

Rationale: High-grade urothelial carcinoma of the bladder is a heterogeneous disease, with molecular subtypes characterized by distinct tumor biologies and prognoses. Our group and others have described the basal and luminal subtypes, and we now report on the discovery of the claudin-low subtype, all of which resemble analogous subtypes in breast cancer. Methods: We analyzed mRNA sequencing and whole exome sequencing data for The Cancer Genome Atlas (TCGA) bladder tumors and tumors collected at UNC. We performed unsupervised hierarchical clustering on relative gene expression values with significance testing using SigClust to identify the claudin-low subtype. Basal, luminal, and claudin-low bladder tumors were compared for enrichment of genetic features (single nucleotide and copy number variation), gene set expression, immune gene signature expression, T cell receptor (TCR) and B cell receptor (BCR) gene segment expression, and number of predicted MHC Class I and Class II neoantigens. We further report on the first use of our VDJician software to reconstruct full-length rearranged BCR sequences from short-read RNA sequencing data in bladder cancer. Results: Claudin-low bladder tumors were defined by low expression of tight junction claudin-proteins, high expression of epithelial-to-mesenchymal transition genes and immune gene signatures, and were associated with reduced overall survival compared to luminal tumors. A minimal gene set classifier to identify claudin-low tumors showed some but not complete overlap with an optimal classifier derived in breast cancer. Claudin-low bladder tumors were enriched for multiple genetic features: increased rates of RB1, EP300, and NCOR1 mutations, increased frequency of EGFR amplification, decreased rates of FGFR3, ELF3, and KDM6A mutations, and decreased frequency of PPARG amplification. Claudin-low tumors showed the highest expression of immune gene signatures (including an immunosuppression signature), however in contrast to basal tumors, increased immune gene signature expression was not associated with prolonged survival. Claudin-low tumors also showed the highest overall expression of rearranged BCR sequences but lowest BCR repertoire diversity. Predicted neoantigen burden did not vary significantly by subtype, however broad cytokine and chemokine expression levels were elevated in claudin-low tumors, potentially related to low PPARG activity driving increased NFKB activity. Conclusions: Claudin-low bladder cancer is a novel molecular subtype with distinct molecular and immunologic features and prognostic significance. Given the presence of dense immune infiltrates, BCR repertoire characteristics consistent with an antigen-driven response, and high expression of immunosuppression genes, claudin-low bladder tumors may be primed to respond to immune checkpoint inhibitor therapy. Citation Format: Benjamin G. Vincent, William Kim, Jordan Kardos, Shengjie Chai, Joel Parker, Lisle Mose, Sara Selitsky, Michael Iglesia, Matthew Milowsky. The novel claudin-low molecular subtype of high-grade urothelial bladder cancer is highly immunogenic yet immunosuppressed. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 5120.


Cancer Research | 2015

Abstract PD1-2: Impact of tumor-infiltrating B-cell clonal diversity on response to neoadjuvant therapy in triple negative and HER2+ breast cancer treated on CALGB (Alliance) 40601 and 40603

Michael D. Iglesia; Benjamin G. Vincent; Jonathan S. Serody; Lisa A. Carey; William T. Barry; William M. Sikov; Clifford A. Hudis; Eric Winer; Charles M. Perou

Background: Tumor infiltrating lymphocytes (TILs) are associated with improved outcomes in breast (BrCa) and ovarian cancer (OvCa). This benefit is largely restricted to the basal-like and HER2-enriched subtypes of BrCa and the immunoreactive subtype of OvCa. It is not known whether TILs respond to a small subset of antigens, similar to an antiviral or antibacterial response, or if the response is nonspecific. We developed a novel method to assess B-cell population diversity by analyzing B-cell receptor (BCR) sequence complexity in mRNA-seq datasets derived from tumor biopsies. B-cells in a subset of basal-like and HER2-enriched BrCa showed high expression of immunoglobulins coinciding with reduced BCR diversity consistent with a restricted epitope-driven immune response. Analysis of DNA patterns from B-cells in basal-like and HER2-enriched BrCa showed a greater prevalence of BCR somatic hypermutation (SHM) suggestive of an antigen-restricted response (Iglesia et al, CCR 2014). Here, we studied the impact of this adaptive immune response on treatment response. Methods: Using two neoadjuvant cooperative group trials in triple-negative (TNBC) and HER2-positive (HER2+) BrCa, we evaluated BCR diversity (as assessed by SHM diversity) as a continuous variable and as a binary variable (diverse/restricted) relative to BCR expression (the Restriction Index) in pre-treatment tumor samples from 265 patients with HER2+ BrCa treated on CALGB 40601, a randomized phase III trial of paclitaxel plus trastuzumab +/- lapatinib, and 443 patients with TNBC treated on CALGB 40603, a randomized phase II trial of standard chemotherapy +/- carboplatin and/or bevacizumab. We examined the relationship between a restricted immune response and pCR rate, the primary endpoint of both studies, overall and within molecular subtypes. Results: In HER2+ BrCa, the combination of high immunoglobulin expression and lower sequence diversity (high Restriction Index) was observed in 28% of the pre-treatment biopsies, and varied by intrinsic subtype, with the greatest prevalence in the HER2-enriched subset (n=80, 46% vs 20% in all others). BCR restriction predicted improved pCR rates in all patients (67% versus 37%, p Conclusions: The presence of a restricted diversity B-cell response in HER2+ breast cancer correlates with improved response to neoadjuvant chemotherapy plus HER2-targeted therapy, which may in part explain its impact on prognosis. We will determine if a similar correlation exists with chemotherapy response in TNBC. This suggests that immunomodulatory therapies supporting a B-cell response may be a promising therapeutic approach to targeting these tumors. Citation Format: Michael D Iglesia, Benjamin G Vincent, Jonathan S Serody, Lisa A Carey, William T Barry, William M Sikov, Clifford A Hudis, Eric M Winer, Charles M Perou. Impact of tumor-infiltrating B-cell clonal diversity on response to neoadjuvant therapy in triple negative and HER2+ breast cancer treated on CALGB (Alliance) 40601 and 40603 [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr PD1-2.


Genome Research | 2012

Tumor-specific retargeting of an oncogenic transcription factor chimera results in dysregulation of chromatin and transcription

Mukund Patel; Jeremy M. Simon; Michael D. Iglesia; Sam B. Wu; Andrew W. McFadden; Jason D. Lieb; Ian J. Davis

Collaboration


Dive into the Michael D. Iglesia's collaboration.

Top Co-Authors

Avatar

Benjamin G. Vincent

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Joel S. Parker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jonathan S. Serody

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Lisa A. Carey

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Katherine A. Hoadley

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Lisle E. Mose

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Sara R. Selitsky

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Anna Rogers

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Bentley R. Midkiff

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge