Skip to Content
Translational Research Program (TRP)
Contact CIP
Show menu
Search this site
Last Updated: 09/24/22

MD Anderson Cancer Center SPORE in Gastrointestinal Cancer

The University of Texas MD Anderson Cancer Center

Principal Investigator(s):

Scott Kopetz, MD, PhD
Scott Kopetz, MD, PhD

Anirban Maitra, MD
Anirban Maitra, MD

Principal Investigators Contact Information

Scott Kopetz, MD, PhD
Professor of Medicine
Department of Gastrointestinal Medical Oncology
The University of Texas MD Anderson Cancer Center
1515 Holcombe Blvd. Unit 426
Houston, TX 77030
(713) 792-2828

Anirban Maitra, MD
Professor and Scientific Director, Sheikh Ahmed Center for Pancreatic Cancer Research
The University of Texas MD Anderson Cancer Center
1515 Holcombe Blvd. #1050
Houston, TX 77030
(713) 745-0861

Overview

CRC is the 2nd most common cause of cancer-related deaths in this country, while PDAC is the 3rd most common cause, underscoring the significance of the work undertaken in this proposal. Our multidisciplinary team is fully engaged in highly innovative translational research including first-in-human trials accelerate the development of therapeutic options available to CRC and PDAC patients.

The MD Anderson SPORE in Gastrointestinal cancer is actively challenging our basic science and clinical investigators to identify and implement leading-edge research that realizes translational impact. Among the current challenges are 1) failure of current immunotherapy agents to improve metastatic CRC outcomes, with the exception of rare subsets representing <5% of patients with microsatellite instability, 2) the slow pace of chemoprevention research and limited repertoire of agents being evaluated in the clinic for populations at-risk for CRC, and 3) necessity to explore novel therapeutic vulnerabilities and eliminate residual disease after standard of care therapy in PDAC. Our SPORE is aggressively meeting each of these challenges by building on strong preclinical data, unique institutional opportunities, exceptional investigators, and proven clinical-translational environment. Led by Drs. Scott Kopetz and Anirban Maitra, this team of investigators with expertise in multidisciplinary translational research and clinical care of CRC and PDAC will achieve these goals by fulfilling the following aims:

Aim 1: To evaluate clinical activity of an optimized combination of immune checkpoint inhibitors with a personalized, neo-antigen peptide vaccine (Project 1).
Aim 2: To determine the contribution of STAT3 signaling to CRC development in high-risk patients with familial syndromes or inflammatory bowel disease (Project 2).
Aim 3: To evaluate metabolic therapeutic vulnerabilities in pancreatic cancer (Project 3).

Our SPORE team is strategically organized to effectively translate preclinical concepts and novel targets rapidly into a clinical setting, with the goal of significant impact on mortality rates from CRC and PDAC.

Project 1: Personalized Adjuvant Immunotherapy for High-risk Colorectal Cancer

Project Co-Leaders:
Michael Overman, MD (Clinical Co-leader)
Gregory Lizee, PhD (Basic Co-leader)
Elizabeth Jaffee, MD (Johns Hopkins) (Clinical Co-leader)

Immunotherapy with antibodies targeting checkpoint blockade molecules PD-1 and CTLA-4 has not demonstrated clinical activity in the vast majority of colorectal cancers (CRC). By contrast, a rare subset of CRC tumors that show microsatellite instability (MSI-high) demonstrate marked responses to PD-1 therapy, similar to or exceeding that observed in other highly mutated cancers such as melanoma and lung adenocarcinoma. This correlation between mutational load and clinical response suggests that the anti-tumor immunity generated by checkpoint inhibitors is mediated largely through the activation of T lymphocytes recognizing mutated peptides presented by HLA class I molecules. While identification of mutated peptide epitope targets in individual patients remains a daunting technical challenge, recent advances in next generation sequencing (NGS) has provided a strong foundation on which to build these efforts. Revealing mutated target antigens in individual cancer patients would facilitate a "precision immunotherapy" strategy in which an immune response against multiple expressed tumor antigens could be generated in patients using multivalent peptide vaccines. The specific objective of this proposal is to generate an effective, personalized vaccine approach for treatment of metastatic CRC patients at high risk of recurrence following surgery. Specifically, we will test the hypothesis that a vaccination strategy targeting multiple mutated CRC tumor antigens along with specific combinations of immune adjuvants will be capable of preventing recurrence in minimal residual disease (MRD) setting in post-hepatectomy CRC patients. As the success of vaccination is likely predicated upon lower volume disease, we propose to investigate our personalized peptide vaccination approach in CRC patients in the adjuvant setting who have detectable mutations in circulating tumor-derived DNA (ctDNA) that can be monitored over time. Our Preliminary Data shows that the proposed personalized vaccination strategy is feasible, as several CRC patients have now undergone vaccination in ongoing clinical trial at UTMDACC. In addition, our pre-clinical mouse data has shown that Toll-like receptor ligands, anti-CD40, and checkpoint blockade can be highly effective combinations of adjuvants in vivo. However, it is critical to understand the optimal combination of agents to use for vaccination in order to translate these findings into more effective vaccine strategies for our CRC patients.

Project 2: Targeting STAT3 to Prevent Colorectal Cancer (CRC) in Hereditary Syndromes and Inflammatory Bowel Disease

Project Co-Leaders:
David Tweardy, MD (Basic Co-leader)
Eduardo Vilar-Sanchez, MD, PhD (Clinical Co-leader)

Evidence is increasing that signal transducer and activator of transcription (STAT) 3 contributes to sporadic and high-risk colorectal cancer (CRC) carcinogenesis arising in a background of inflammation such as inflammatory bowel disease (IBD), and also hereditary populations such as familial adenomatous polyposis (FAP) and Lynch syndrome (LS). IBD, which presents as either ulcerative colitis (UC) or Crohn's disease (CD), constitutes attractive models to study contribution of inflammation to colorectal carcinogenesis. STAT3 is expressed in two isoforms—STAT3α and STAT3β. STAT3α is proinflammatory and anti-apoptotic, while STAT3β antagonizes these effects of STAT3α. IBD in mouse models induced by either dextran sodium salt (DSS;UC model) or trinitrobenzoic acid (TNBS;CD model) was more severe in transgenic mice expressing only STAT3α compared to wild type mice. We developed a potent small-molecule STAT3 inhibitor, TTI-101 (formerly C188-9), that targets the phosphotyrosyl peptide-binding pocket within the STAT3 SH2 domain, which blocks STAT3 activation [phosphorylation on tyrosine (Y) 705, pY-STAT3]. TTI-101 administration prevented IBD caused by both DSS and TNBS. In other studies, mice with STAT3-deficient intestinal epithelial cells demonstrated reduced tumor size and reduced tumor incidence in a model of colitis-associated CRC [azoxymethane (AM) plus DSS]. FAP is caused by germline mutations in APC. Genetically reducing levels of STAT3 in Apc GEMM decreased the number of intestinal polyps. STAT3 activation results in extra-nuclear sequestration of MSH3, which may further impair DNA mismatch repair (dMMR) in LS enterocytes bearing a mutation in other dMMR enzymes resulting in increased CRC risk. Our preliminary data using a CLIA-certified pY-STAT3 IHC stain and scoring system supports activation of STAT3 signaling in normal-appearing mucosa that is further increased in CRC samples from IBD, FAP and LS patients. The long-term goal of Project 2 is to determine if TTI-101 will benefit in CRC prevention. The central hypotheses are: STAT3 contributes to CRC development in patients at risk for CRC and can be targeted successfully with TTI-101. The objectives are: determine the effects of targeting STAT3 with TTI-101 for CRC prevention in mouse models and the contribution of STAT3 signaling to CRC development in IBD, FAP, and LS patients.

Project 3: Inhibiting Oxidative Phosphorylation in Pancreatic Cancer

Project Co-Leaders:
Shubham Pant, MD (Clinical Co-leader)
Giulio Draetta, MD, PhD (Basic Co-leader)

Despite a better understanding of the genomic landscape and the importance of the tumor microenvironment, there has been no meaningful shift in the overall survival for PDAC. Thus, new therapeutic strategies for this deadly disease represent an urgent unmet clinical need. Over 90% of PDAC harbor activating KRAS mutations, which are an early event in disease pathogenesis as they are present in pancreatic intraepithelial neoplasias (PanINs), the precursor lesion for PDAC. Increasingly, inter- and intra-tumoral heterogeneity in PDAC are appreciated, including KRAS dependency. Differential dependence on KRAS been linked with altered metabolic dependencies. In one specific context, our group used an inducible oncogenic Kras-driven PDAC model to demonstrate that, upon extinction of oncogenic Kras, the persistent Kras-independent tumor-initiating cells (TICs) exhibited a metabolic profile very different from that of the Kras-dependent cancer cells of the bulk tumor. Whereas the Kras-dependent cancer cells of the bulk tumor exhibited high levels of glycolysis and metabolic dependencies, the Kras-independent TICs showed impaired glycolysis and increased mitochondrial respiration. Similar observations were made in TICs derived from human PDAC PDX models, which exhibited decreased glucose flux through glycolysis and elevated OXPHOS activity. These TICs harbor limited metabolic plasticity, rendering them particularly sensitive to inhibition of mitochondrial activity. Thus, heterogeneity of PDAC is not only defined on the genomic and cellular levels, but also defined by distinctive metabolism programs controlled by oncogenic signaling. However, to date, the documented dependency of some tumors or tumor cell subpopulations on OXPHOS has not yet been exploited therapeutically. We will explore the biology of response to treatment with metabolic inhibitors in these contexts, using a combination of ex vivo and in vivo studies. We will also evaluate safety and patient response (via clinical correlatives including transcriptomic signatures, hyperpolarized pyruvate-magnetic resonance imaging, quantitative CT scan) in a phase 1b clinical trial of IM156, a novel OXPHOS inhibitor, in combination with gemcitabine and nab-paclitaxel in patients with treatment-naïve or refractory disease (phase 1b). The trial protocol is FDA- and IRB-approved, and enrollment is set to commence in September 2022.

Administrative Core

Core Directors:
Scott Kopetz, MD, PhD
Anirban Maitra, MBBS

The Administrative Core will be responsible for the successful execution and management of all SPORE activities related to financial oversight and coordination, organization of all necessary meetings, and publicity and record keeping for all projects and the two other cores. This group will also provide regulatory oversight activities for clinical trials; ensure compliance with all institutional, federal, and NCI-specific regulations; and oversee the peer-review and oversight processes of the Career Enhancement Program and Developmental Research Program. The core will be led by Drs. Scott Kopetz, Anirban Maitra and David Menter.

Biospecimen and Pathology Core

Core Directors:
Dipen Maru, MD
Huamin Wang, MD, PhD

Core 1 will coordinate efforts related to collection, processing, storage and distribution of annotated human and murine biospecimens for all of the SPORE projects, including the Career Enhancement Program (CEP) and Developmental Research Program (DRP). The Core will be co-led by two internationally reputed gastrointestinal/pancreatic pathologists, Drs. Dipen Maru and Huamin Wang. For human biospecimens, the core will interface with various Institutional systems (ePRTCL, PDMS, CORe, Electronic Health Record [EHR], or the Biospecimen Information Management System [BIMS]). Biospecimen resources from the lower gastrointestinal tissue bank include freshly collected/snap frozen and formalin fixed paraffin embedded tumor and normal specimens from more than 2500 resected hepatic colorectal metastases (including TMAs), freshly collected and snap frozen adenomas from 334 patients and formalin fixed paraffin embedded specimens from 870 or more patients with sporadic adenoma or familial adenomatous polyposis. Existing biospecimen resources available in the pancreatic bank include freshly collected/snap frozen tumor and normal tissue sample from Whipple resection for pancreatic ductal adenocarcinoma from 232 patients, with formalin fixed paraffin embedded specimens and additional 672 patients, including tissue microarrays from pancreatic ductal adenocarcinoma and intraductal pancreatic mucinous neoplasms. The Core will support Project 1 by coordinating prospective blood collection, cryopreservation and transport with the ITB. Specifically, Core faculty will prospectively collect, process and distribute fresh tumor and normal samples from hepatic colorectal metastases after obtaining mirror image section for histology quality control. The Core will provide formalin fixed paraffin embedded samples of normal, adenoma and carcinoma to Project 2. The Core will also provide biospecimen qualification services, including but not limited to, histopathologic characterization of human and murine tissues treated with STAT3 inhibitor, and immunohistochemistry staining and interpretation, including validation of p-STAT3 staining by automated image analysis in a CLIA-certified facility. The Core will provide freshly resected PDAC samples for patient derived xenografts and ex vivo live tissue sensitivity assay (LTSA) for Project 3. In addition, the Core will provide histopathology characterization, immunohistochemistry services and interpretation guidelines for both preclinical samples from the ongoing co-clinical trials. The Core personnel, along with the ITB, will enter detailed information related to all processes of biospecimen collection, processing, qualification, distribution and analytes extraction into BIMS. The Core activities will lead to enhancement of these functionalities of Tissue Station and design a new interface specific for GI SPORE in the Tissue Station.

Biostatistics and Bioinformatics Core

Core Directors:
Ying Yuan, MD
Ryan Sun, PhD

The Biostatistics and Bioinformatics (Core 2) provides comprehensive service to guide design of experiments, to optimize quantitative data analysis, and to maintain statistical justification and interpretation of results. Specifically, Core 2 will implement sound experimental design principles that are tailored to address specific scientific questions for each project. Core 2 will also carry out data analyses using suitable statistical methods and bioinformatics algorithms, and will contribute to the interpretation of results through written reports and frequent interactions with project investigators. Whenever appropriate, Core 2 will develop new analysis tools to address new challenges in the analysis of various data, especially high-throughput genomic and proteomic data. Members of Core 2 will participate in monthly SPORE meetings with all project investigators, ensuring that statistical and data analysis/management issues are carefully considered during all phases of each SPORE experiment. Thus, from inception to reporting and publication, basic laboratory and translational experiments will benefit from the SPORE program that will be used to augment existing MD Anderson Cancer Center biostatistics resources.

Developmental Research Program

Program Directors:
Scott Kopetz, MD, PhD
Anirban Maitra, MBBS

The purpose of the Developmental Research Program (DRP) is to fund highly innovative translational studies in colorectal cancer (CRC) or pancreatic cancer (PDAC) that currently lack a human endpoint as mandated by the SPORE guidelines, but that could become full SPORE projects with requisite DRP support, or compete successfully for funding outside of the SPORE mechanism. The DRP provides a unique venue for making available critical financial support, as well as disease-specific intellectual and resource expertise, through a program that is rapidly responsive to new ideas or initiatives, including from investigators whose current work may not focus exclusively in CRC or PDAC research. This process will therefore be a major educational activity that is further anticipated to stimulate translational research in CRC or PDAC and encourage the participation of both basic science researchers and clinical investigators in translational research.

Career Enhancement Program

Program Directors:
Scott Kopetz, MD, PhD
Elizabeth Jaffee, MD (Johns Hopkins)

The Career Enhancement Program (CEP) is designed to identify, recruit, and mentor junior scientists, including underrepresented investigators (minorities and women), for performing translational research in pancreatic (PDAC) and colorectal cancer (CRC). Fellowship training for physicians in gastrointestinal oncology and gastrointestinal pathology usually emphasizes clinical care and clinical trials. Both clinical and laboratory-based researchers are eligible for funding providing that they are proposing projects that are translational in nature.

Institutional SPORE Website

https://www.mdanderson.org/research/departments-labs-institutes/spores/gastrointestinal-cancer-spore.html