31 results on '"Christina Wu"'
Search Results
2. supplemental legend from Systemic Immune Activity Predicts Overall Survival in Treatment-Naïve Patients with Metastatic Pancreatic Cancer
- Author
-
Gregory B. Lesinski, Tanios Bekaii-Saab, Miguel Villalona-Calero, Anne M. Noonan, Daniel Ahn, Sanaa Tahiri, Kristen Ciombor, Christina Wu, Sameh Mikhail, Susan Geyer, Thomas A. Mace, and Matthew R. Farren
- Abstract
supplemental legend
- Published
- 2023
3. Data from Systemic Immune Activity Predicts Overall Survival in Treatment-Naïve Patients with Metastatic Pancreatic Cancer
- Author
-
Gregory B. Lesinski, Tanios Bekaii-Saab, Miguel Villalona-Calero, Anne M. Noonan, Daniel Ahn, Sanaa Tahiri, Kristen Ciombor, Christina Wu, Sameh Mikhail, Susan Geyer, Thomas A. Mace, and Matthew R. Farren
- Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with a 5-year survival rate Experimental Design: Peripheral blood was collected from 73 patients presenting with previously untreated metastatic PDAC. Extensive immunologic profiling was conducted to assess relationships between OS and the level of soluble plasma biomarkers or detailed immune cell phenotypes as measured by flow cytometry.Results: Higher baseline levels of the immunosuppressive cytokines IL6 and IL10 were strongly associated with poorer OS (P = 0.008 and 0.026, respectively; HR = 1.16 and 1.28, respectively), whereas higher levels of the monocyte chemoattractant MCP-1 were associated with significantly longer OS (P = 0.045; HR = 0.69). Patients with a greater proportion of antigen-experienced T cells (CD45RO+) had longer OS (CD4 P = 0.032; CD8 P = 0.036; HR = 0.36 and 0.61, respectively). Although greater expression of the T-cell checkpoint molecule CTLA-4 on CD8+ T cells was associated with significantly shorter OS (P = 0.020; HR = 1.53), the TIM3 molecule had a positive association with survival when expressed on CD4+ T cells (P = 0.046; HR = 0.62).Conclusions: These data support the hypothesis that baseline immune status predicts PDAC disease course and overall patient survival. To our knowledge, this work represents the largest cohort and most comprehensive immune profiling of treatment-naïve metastatic PDAC patients to date. Clin Cancer Res; 22(10); 2565–74. ©2015 AACR.
- Published
- 2023
4. Data from Phase I Immunotherapy Trial with Two Chimeric HER-2 B-Cell Peptide Vaccines Emulsified in Montanide ISA 720VG and Nor-MDP Adjuvant in Patients with Advanced Solid Tumors
- Author
-
Pravin T.P. Kaumaya, Jay Overholser, Lai Wei, Jeffrey Fowler, Bhuvaneswari Ramaswamy, Maryam Lustberg, Amir Mortazavi, Christina Wu, Daniel H. Ahn, Robert Wesolowski, and Tanios Bekaii-Saab
- Abstract
Purpose:This first-in-human phase I study (NCT 01417546) evaluated the safety profile, optimal immunologic/biological dose (OID/OBD), and immunogenicity of the combination of two peptide B-cell epitope vaccines engineered to represent the trastuzumab- and pertuzumab-binding sites. Although trastuzumab and pertuzumab have been approved for clinical use, patients often develop resistance to these therapies. We have advanced a new paradigm in immunotherapy that focuses on humoral responses based on conformational B-cell epitope vaccines.Patients and Methods:The vaccine is comprised of two chimeric HER-2 B-cell peptide vaccines incorporating a “promiscuous T-cell epitope.” Patients were immunized with the vaccine constructs emulsified with nor-muramyl-dipeptide adjuvant in a water-in-oil Montanide ISA 720VG vehicle. Eligible patients with metastatic and/or recurrent solid tumors received three inoculations every 3 weeks.Results:Forty-nine patients with a median of 4 prior lines of chemotherapy received at least 1 vaccination. Twenty-eight patients completed the 3 vaccination regimens. Six patients received 1 six-month boost after the regimen, and one patient received 7 six-month boosts. No serious adverse reactions or dose-limiting toxicities were observed. The vaccine was well tolerated with dose level 2 as the recommended phase II dose. The most common related toxicity in all patients was injection-site reactions (24%). Two patients had a partial response, 14 had stable disease, and 19 had progressive disease.Conclusions:The study vaccine is safe, exhibits antitumor activity, and shows preliminary indication that peptide vaccination may avoid therapeutic resistance and offer a promising alternative to monoclonal antibody therapies.
- Published
- 2023
5. Supplementary fig 1 from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Supplementary fig 1 Flowchart for trial design
- Published
- 2023
6. Supplementary fig 2 from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Supplementary fig 2 Schema for trial design
- Published
- 2023
7. Supplementary Materials from Systemic Immune Activity Predicts Overall Survival in Treatment-Naïve Patients with Metastatic Pancreatic Cancer
- Author
-
Gregory B. Lesinski, Tanios Bekaii-Saab, Miguel Villalona-Calero, Anne M. Noonan, Daniel Ahn, Sanaa Tahiri, Kristen Ciombor, Christina Wu, Sameh Mikhail, Susan Geyer, Thomas A. Mace, and Matthew R. Farren
- Abstract
Table S1. Antibodies used for flow cytometry Table S2. Monte Carlo cross-validation of continuous measure markers. Table S3. Monte Carlo cross-validation of dichotomous measure markers. Table S4. Pancreatic cancer patient circulating immune cell frequencies Figure S1. Overall survival does not predict plasma MCP-1 levels
- Published
- 2023
8. Supplementary Data from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Table showing next generation sequencing of tumor samples
- Published
- 2023
9. Supplementary Data from Phase I Immunotherapy Trial with Two Chimeric HER-2 B-Cell Peptide Vaccines Emulsified in Montanide ISA 720VG and Nor-MDP Adjuvant in Patients with Advanced Solid Tumors
- Author
-
Pravin T.P. Kaumaya, Jay Overholser, Lai Wei, Jeffrey Fowler, Bhuvaneswari Ramaswamy, Maryam Lustberg, Amir Mortazavi, Christina Wu, Daniel H. Ahn, Robert Wesolowski, and Tanios Bekaii-Saab
- Abstract
This file contains supplemental materials, tables, and figures.
- Published
- 2023
10. Supplementary fig 5 from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Supplementary fig 5 Degree of Reduction (%) in Tumor pERK Levels From Baseline to After Trametinib Monotherapy and Correlation With pCR
- Published
- 2023
11. Supplementary fig 4 from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Supplementary fig 4 Baseline p-ERK score in Tumor Tissue Correlated to RAS/RAF Mutation Status
- Published
- 2023
12. Data from Phase I Trial of Trametinib with Neoadjuvant Chemoradiation in Patients with Locally Advanced Rectal Cancer
- Author
-
Evan Wuthrick, Tanios Bekaii-Saab, Sameek Roychowdhury, Sherif Abdel-Misih, Alan Harzman, Mark Arnold, Somashekar G. Krishna, Cynthia Timmers, Dana B. Cardin, Kristen K. Ciombor, Sameh Mikhail, Wei Chen, Lai Wei, Amy Webb, Ryan Robb, Terence M. Williams, and Christina Wu
- Abstract
Purpose:The RAS/RAF/MEK/ERK signaling pathway is critical to the development of colorectal cancers, and KRAS, NRAS, and BRAF mutations foster resistance to radiation. We performed a phase I trial to determine the safety of trametinib, a potent MEK1/2 inhibitor, with 5-fluorouracil (5-FU) chemoradiation therapy (CRT) in patients with locally advanced rectal cancer (LARC).Patients and Methods:Patients with stage II/III rectal cancer were enrolled on a phase I study with 3+3 study design, with an expansion cohort of 9 patients at the MTD. Following a 5-day trametinib lead-in, with pre- and posttreatment tumor biopsies, patients received trametinib and CRT, surgery, and adjuvant chemotherapy. Trametinib was given orally daily at 3 dose levels: 0.5 mg, 1 mg, and 2 mg. CRT consisted of infusional 5-FU 225 mg/m2/day and radiation dose of 28 daily fractions of 1.8 Gy (total 50.4 Gy). The primary endpoint was to identify the MTD and recommended phase II dose. IHC staining for phosphorylated ERK (pERK) and genomic profiling was performed on the tumor samples.Results:Patients were enrolled to all dose levels, and 18 patients were evaluable for toxicities and responses. Treatment was well tolerated, and there was one dose-limiting toxicity of diarrhea, which was attributed to CRT rather than trametinib. At the 2 mg dose level, 25% had pathologic complete response. IHC staining confirmed dose-dependent decrease in pERK with increasing trametinib doses.Conclusions:The combination of trametinib with 5-FU CRT is safe and well tolerated, and may warrant additional study in a phase II trial, perhaps in a RAS/RAF-mutant selected population.
- Published
- 2023
13. A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors
- Author
-
Suresh S. Ramalingam, Conor E. Steuer, Mehmet Asim Bilen, Hannah Collins, Sagar Lonial, Zhengjia Chen, Bradley C. Carthon, Mehmet Akce, Olatunji B. Alese, Taofeek K. Owonikoko, Christina Wu, Ragini R Kudchagkar, David H. Lawson, R. Donald Harvey, Colleen Lewis, Chao Zhang, Bassel F. El-Rayes, Wayne Harris, Walid L. Shaib, Gabriel Sica, and Fadlo R. Khuri
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Nausea ,Morpholines ,Buparlisib ,Aminopyridines ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Mucositis ,Humans ,Tissue Distribution ,Everolimus ,Survival rate ,Aged ,business.industry ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Female ,medicine.symptom ,business ,Progressive disease ,Follow-Up Studies ,medicine.drug - Abstract
Purpose: Concurrent inhibition of mTOR and PI3K led to improved efficacy in preclinical models and provided the rationale for this phase I study of everolimus and buparlisib (BKM120) in patients with advanced solid tumor. Patients and Methods: We used the Bayesian Escalation with Overdose Control design to test escalating doses of everolimus (5 or 10 mg) and buparlisib (20, 40, 60, 80, and 100 mg) in eligible patients. Pharmacokinetic assessment was conducted using blood samples collected on cycle 1, days 8 and 15. Pharmacodynamic impact on mTOR/PI3K pathway modulation evaluated in paired skin biopsies collected at baseline and end of cycle 1. Results: We enrolled 43 patients, median age of 63 (range, 39–78) years; 25 (58.1%) females, 35 (81.4%) Caucasians, and 8 (18.6%) Blacks. The most frequent toxicities were hyperglycemia, diarrhea, nausea, fatigue, and aspartate aminotransferase elevation. Dose-limiting toxicities observed in 7 patients were fatigue (3), hyperglycemia (2), mucositis (1), acute kidney injury (1), and urinary tract infection (1). The recommended phase II dose (RP2D) for the combination was established as everolimus (5 mg) and buparlisib (60 mg). The best response in 27 evaluable patients was progressive disease and stable disease in 3 (11%) and 24 (89%), respectively. The median progression-free survival and overall survival were 2.7 (1.8–4.2) and 9 (6.4–13.2) months. Steady-state pharmacokinetic analysis showed dose-normalized maximum concentrations and AUC values for everolimus and buparlisib in combination to be comparable with single-agent pharmacokinetic. Conclusions: The combination of everolimus and buparlisib is safe and well-tolerated at the RP2D of 5 and 60 mg on a continuous daily schedule.
- Published
- 2020
14. Phase I Immunotherapy Trial with Two Chimeric HER-2 B-Cell Peptide Vaccines Emulsified in Montanide ISA 720VG and Nor-MDP Adjuvant in Patients with Advanced Solid Tumors
- Author
-
Robert Wesolowski, Maryam B. Lustberg, Jay Overholser, Bhuvaneswari Ramaswamy, Christina Wu, Tanios Bekaii-Saab, Pravin T. P. Kaumaya, Lai Wei, Daniel H. Ahn, Amir Mortazavi, and Jeffrey M. Fowler
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Receptor, ErbB-2 ,medicine.medical_treatment ,Oleic Acids ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Adjuvants, Immunologic ,Trastuzumab ,Neoplasms ,Internal medicine ,Tumor Cells, Cultured ,medicine ,Humans ,Mannitol ,Aged ,Cell Proliferation ,Aged, 80 and over ,B-Lymphocytes ,business.industry ,Immunogenicity ,Immunotherapy ,Middle Aged ,medicine.disease ,Vaccination ,Regimen ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Vaccines, Subunit ,Epitopes, B-Lymphocyte ,Female ,Immunization ,Pertuzumab ,business ,Adjuvant ,Progressive disease ,medicine.drug - Abstract
Purpose: This first-in-human phase I study (NCT 01417546) evaluated the safety profile, optimal immunologic/biological dose (OID/OBD), and immunogenicity of the combination of two peptide B-cell epitope vaccines engineered to represent the trastuzumab- and pertuzumab-binding sites. Although trastuzumab and pertuzumab have been approved for clinical use, patients often develop resistance to these therapies. We have advanced a new paradigm in immunotherapy that focuses on humoral responses based on conformational B-cell epitope vaccines. Patients and Methods: The vaccine is comprised of two chimeric HER-2 B-cell peptide vaccines incorporating a “promiscuous T-cell epitope.” Patients were immunized with the vaccine constructs emulsified with nor-muramyl-dipeptide adjuvant in a water-in-oil Montanide ISA 720VG vehicle. Eligible patients with metastatic and/or recurrent solid tumors received three inoculations every 3 weeks. Results: Forty-nine patients with a median of 4 prior lines of chemotherapy received at least 1 vaccination. Twenty-eight patients completed the 3 vaccination regimens. Six patients received 1 six-month boost after the regimen, and one patient received 7 six-month boosts. No serious adverse reactions or dose-limiting toxicities were observed. The vaccine was well tolerated with dose level 2 as the recommended phase II dose. The most common related toxicity in all patients was injection-site reactions (24%). Two patients had a partial response, 14 had stable disease, and 19 had progressive disease. Conclusions: The study vaccine is safe, exhibits antitumor activity, and shows preliminary indication that peptide vaccination may avoid therapeutic resistance and offer a promising alternative to monoclonal antibody therapies.
- Published
- 2019
15. Abstract 5790: Genomic landscape of circulating tumor DNA alterations in patients with paraganglioma and pheochromocytoma
- Author
-
Lana Khalil, Jill Tsai, Leylah Drusbosky, Olatunji Alese, Maria Diab, Mehmet Akce, Christina Wu, Olumide Babjide Gbolahan, Bassel El-Rayes, and Walid Shaib
- Subjects
Cancer Research ,Oncology - Abstract
Background: Paragangliomas (PGLs) and Pheochromocytomas (PCCs) are rare neuroendocrine tumors (NETs). PGLs arise from chromaffin cells in the ganglia of the autonomic nervous system, while PCCs arise from chromaffin cells in the adrenal medulla. The genomic landscapes of PGLs and PCCs have not been well studied. Thus, the aim of this study is to report differences of mutational occurrences and confirm the feasibility of next generation sequencing (NGS) testing circulating tumor DNA (ctDNA) in patients with PGL and PCC. Patients and Methods: Molecular alterations in 46 plasma samples were evaluated using a commercially available ctDNA assay (Guardant360® or Guardant360® CDx) across multiple institutions from 2016-2021. The tests detect single nucleotide variants and indels in 54-83 genes with copy number amplifications and fusions in selected genes. Results: Of the 24 PGL patients, there was a median age of 55 (range:28-78) years and 14 (58%) patients were male. Of the 22 PCC patients, there was a median age of 56 (range:28-86) years and 12 (54.5%) patients were male. Genetic alterations were identified in 16 (67%) PGL and 17 (77%) PCC patients. Of the 16 PGL samples with alterations, TP53 associated genes were most commonly altered (44%), followed by ATM (25%), FGFR2 (19%), APC (13%), BRAF (13%), BRCA1 (13%), CCND2 (13%), FGFR3 (13%), IDH2 (13%), KRAS (13%), PDGFRA (13%), RB1 (13%), TERT(13%), ALK (6%), ARID1A (6%), BRCA2 (6%), CCND1 (6%), CDK6 (6%), CDK12 (6%), EGFR (6%), FGFR1 (6%), KIT (6%), MET (6%), NF1 (6%), NRAS (6%), PIK3CA (6%), PTEN (6%), and ROS1 (6%). Of the 17 PCC samples with alterations, TP53 was most commonly altered (41%), followed by ATM (35%), NF1 (24%), FGFR1 (18%), APC (13%), EGFR (12%), MET (12%), MYC (12%), NOTCH1 (12%), PDGFRA (12%), TSC1 (12%), AR (6%), ARID1A(13%), BRAF (6%), BRCA1 (6%), BRCA2 (6%), CCND1 (6%), CDK6 (6%), CHEK2 (6%), ERBB2 (6%), EZH2 (6%), FGFR2 (6%), IDH2 (6%), KIT (6%), KRAS (6%), NRAS (6%), NTRK1 (6%), NTRK2 (6%), and VHL (6%). 21% of PGL and 41% of PCC patients reported alterations associated with therapies approved in other indications including genes in the MAPK pathway and the homologous recombination repair pathway. Conclusions: Liquid biopsy is a non-invasive method that can provide personalized treatment options for patients. In this study, we found that evaluation of ctDNA was feasible among individuals with advanced PGL and PCC. We report a high rate of homologous recombinant deficiencies that are in need of evaluation in future Percentage refers to frequency of patients with an alteration detected. Citation Format: Lana Khalil, Jill Tsai, Leylah Drusbosky, Olatunji Alese, Maria Diab, Mehmet Akce, Christina Wu, Olumide Babjide Gbolahan, Bassel El-Rayes, Walid Shaib. Genomic landscape of circulating tumor DNA alterations in patients with paraganglioma and pheochromocytoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5790.
- Published
- 2022
16. Abstract 3482: Correlative analysis of metformin and nivolumab combination in treatment-refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC)
- Author
-
Batoul Farran, Jeffrey M. Switchenko, Lana Khalil, Walid L. Shaib, Brian Olson, Amanda Ruggieri, Christina Wu, Olatunji B. Alese, Maria Diab, Gregory B. Lesinski, Bassel El-Rayes, and Mehmet Akce
- Subjects
Cancer Research ,Oncology - Abstract
Background: Preclinical results indicate that metformin can modulate immune cell populations in the tumor microenvironment of solid tumors by diminishing exhaustion of CD8+ tumor infiltrating cells and improving T cell responses. Studies also suggest that metformin could complement PD-1 blockade and potentiate its antitumor activity. Previously we reported the results of a phase II trial with metformin and nivolumab and here we report the findings of correlative analysis of the prospectively collected research samples of 18 patients. Methods: We conducted a phase II trial with nivolumab and metformin in treatment-refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC). Nivolumab 480 mg IV every 4 weeks and metformin 1000 mg orally twice daily was administered in 28-day cycles following a 14-day metformin only lead-in phase. The primary endpoint was overall response rate (ORR). Secondary endpoints were overall survival (OS) and progression free survival (PFS). Pre-treatment and on-treatment research biopsies and correlative peripheral blood specimens were collected. Paired biopsies obtained at baseline and following treatment with metformin only (n=9) or metformin and nivolumab (n=9) and were stained with a panel of 13 markers using ChipCytometry technology by Canopy Biosciences. Sample was assessed prior to establishing the multiplex assay. 30 out of 36 samples were imaged and analyzed up to 30 Fields of View. Single cell recognition and quantitative biomarker analysis were performed to compare immune cell numbers and population distribution in pre- versus post-treatment samples. Results: As previously reported, no patients had objective response based on RECIST version 1.1 and the study was stopped after the first stage for futility. Median OS and PFS was 5.1 months [95% CI (2-11.7)] and 2.3 months [95% CI (1.7-2.4)], respectively. Multiplex analysis of tissues from patients receiving lead in with metformin alone revealed fewer effector CD4 T cells and effector and effector memory CD8 T cells after treatment vs. baseline biopsy. Biopsy tissue from patients treated with metformin and nivolumab had lower pAMPK and decreased PDL-1 expression vs. baseline. The combination also increased percentages of leukocytes, effector CD4 T cells, effector and effector memory CD8 T cells as well as levels of PDL1-Tim3+ cells. Conclusion: In the setting of MSS mCRC, metformin as a single agent did not enhance effector CD4 and CD8 T cell percentages in clinical samples in our patient cohort. Metformin in combination with nivolumab was associated with increased percentages of effector CD4 and CD8 T cells in biopsy specimens, although these improvements did not translate into enhanced clinical endpoints. Analysis of peripheral blood samples are currently underway to corroborate the findings in the tissue samples. Citation Format: Batoul Farran, Jeffrey M. Switchenko, Lana Khalil, Walid L. Shaib, Brian Olson, Amanda Ruggieri, Christina Wu, Olatunji B. Alese, Maria Diab, Gregory B. Lesinski, Bassel El-Rayes, Mehmet Akce. Correlative analysis of metformin and nivolumab combination in treatment-refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3482.
- Published
- 2022
17. Abstract A15: Integrated biomarker trials to evaluate myeloid and lymphoid composition of HNSCC and solid tumors treated with pepinemab and combinations with checkpoint inhibitors
- Author
-
Michael C. Lowe, Clint T. Allen, Nabil F. Saba, Christina Wu, Ernest S. Smith, Conor E. Steuer, John E. Leonard, Paul E. Clavijo, Gregory B. Lesinski, Desa Rae Pastore, Brian Olson, Elizabeth E. Evans, Crystal Mallow, Ragini R. Kudchadkar, Terrence L. Fisher, and Maurice Zauderer
- Subjects
Cancer Research ,Tumor microenvironment ,Myeloid ,business.industry ,Colorectal cancer ,Cancer ,Ipilimumab ,medicine.disease ,Head and neck squamous-cell carcinoma ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Biomarker (medicine) ,Nivolumab ,business ,medicine.drug - Abstract
Recruitment of immunosuppressive myeloid cells into the tumor microenvironment (TME) is a critical limitation to the efficacy of immune checkpoint inhibitors (ICIs) in patients with head and neck squamous cell carcinoma (HNSCC). In preclinical models, antibody blockade of Semaphorin 4D (SEMA4D, CD100) reduced function and recruitment of immunosuppressive myeloid cells within the TME. Importantly, combinations of anti-SEMA4D with ICIs enhanced T-cell activity and tumor regression. Pepinemab, an IgG4 humanized monoclonal antibody targeting SEMA4D, is currently being evaluated in window of opportunity, integrated biomarker trials to characterize immunomodulatory effects of treatment. SEMA4D exerts multifaceted effects within the tumor microenvironment by creating a barrier at the tumor-stroma margin, restricting immune cell infiltration and promoting immunosuppressive activity of myeloid-derived cells. In preclinical in vitro and in vivo studies, blocking antibody to SEMA4D directly enhanced M1/M2 ratio and reduced both expression of chemokines that recruit MDSC and the ability of MDSC to suppress T-cell activity. SEMA4D antibody treatment simultaneously restored the ability of dendritic cells and cytotoxic T cells to infiltrate the TME and increased ratio of Teffector to Tregulatory cells. This coordinated shift from immunosuppression to tumoricidal activity complemented effects of other immunotherapies in syngeneic tumor models. At present, three biomarker-driven window of opportunity trials are recruiting patients with four resectable indications to investigate novel combinations of pepinemab with ICIs: 1) HNSCC (NCT03690986, n=36), 2) pancreatic ductal adenocarcinoma and colorectal cancer with resectable liver metastases (NCT03373188, n=32), and 3) metastatic melanoma (NCT03769155, n=36). Presurgical treatment cohorts include combinations of pepinemab with nivolumab and /or ipilimumab, single agents, or no treatment. Three to seven weeks later, surgically resected tumors are collected under the guidance of a pathologist for comparison of tumor infiltrates across treatment groups and with a predose tissue biopsy. Blood is collected for PK, PD, and additional correlative biomarker assessments. Correlative multiplex flow cytometric and immunohistochemistry panels have been established to phenotype cells in the TME and periphery; preliminary biomarker analysis will be presented. Additional objectives include evaluation of pathologic response and extension of the previously reported safety profile of pepinemab to additional ICI combination therapies. Twelve subjects, including two HNSCC patients, have been enrolled in these studies as of 01 Feb 2019. These trials will provide the first integrated clinical assessment of the use of anti-SEMA4D antibody to reprogram the TME. Citation Format: Elizabeth E. Evans, Gregory B. Lesinski, Terrence L. Fisher, Crystal Mallow, Brian Olson, Paul E. Clavijo, John E. Leonard, Desa Rae Pastore, Ernest S. Smith, Maurice Zauderer, Clint T. Allen, Michael Lowe, Ragini Kudchadkar, Christina Wu, Conor Steuer, Nabil F. Saba. Integrated biomarker trials to evaluate myeloid and lymphoid composition of HNSCC and solid tumors treated with pepinemab and combinations with checkpoint inhibitors [abstract]. In: Proceedings of the AACR-AHNS Head and Neck Cancer Conference: Optimizing Survival and Quality of Life through Basic, Clinical, and Translational Research; 2019 Apr 29-30; Austin, TX. Philadelphia (PA): AACR; Clin Cancer Res 2020;26(12_Suppl_2):Abstract nr A15.
- Published
- 2020
18. Abstract B48: Targeting the semaphorin 4D-plexin B axis to augment FOLFIRINOX in a murine model of pancreatic adenocarcinoma
- Author
-
Luis I. Ruffolo, Maurice Zauderer, Brian Olson, Gregory B. Lesinski, Alexander C. Chacon, Mary A. Georger, Brian A. Belt, Elizabeth E. Evans, David C. Linehan, Crystal Mallow, Katherine M. Jackson, Paul R. Burchard, Rachel Jewell, Peter A. Prieto, Christina Wu, Terrence L. Fisher, Nicholas A. Ullman, and Alexa Melucci
- Subjects
Cancer Research ,Tumor microenvironment ,Myeloid ,biology ,business.industry ,FOLFIRINOX ,Plexin ,SEMA4D ,medicine.disease ,Immune checkpoint ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,biology.protein ,Cancer research ,Adenocarcinoma ,business - Abstract
Background: Pancreatic adenocarcinoma (PDAC) is the fourth leading cause of cancer-related mortality, with dismal 5-year prognosis of 8%, in part due to poor response to available therapies. Thus, new systemic cancer control therapies are in dire need. Semaphorin 4D (SEMA4D) is a soluble and membrane bound glycoprotein that binds its cognate Plexin B1/B2 receptors, expressed on monocytic and granulocytic leukocytes. Prior work has shown that increased expression of SEMA4D and Plexin B in resected PDAC patients is associated with lymph node and distant metastasis, as well as a worse prognosis. Additionally, SEMA4D blockade has conferred improved immune checkpoint blockade response in murine models of colorectal carcinoma and head and neck squamous cell carcinoma via abrogation of myeloid-derived suppressor cell recruitment and function, as well as enhanced T-cell recruitment and activity within the tumor microenvironment. Here we study the effects of SEMA4D blockade in a murine model of PDAC. Methods: C57b/6 mice were orthotopically injected with murine PDAC line (KP2) derived from KRASG12D,TP53Flox/Wt;P48-Cre autochthonous tumors. Mice were treated with Folfirinox (5-FU, irinotecan, oxaliplatin, weekly), immune checkpoint blockade (ICB) (anti-PD1, anti-CTLA-4 mAbs bi-weekly), and anti-SEMA4D mAB (biweekly). Peripheral blood and tumor-infiltrating leukocytes from patients with PDAC undergoing pancreaticoduodenectomy were assessed for Plexin B1 via flow cytometry. Results: Human PDAC demonstrates penetration of Plexin B1 positive leukocytes, most notably tumor-associated macrophages, neutrophils, and monocytes. Mice injected with KP2 developed tumors detectable via high-frequency ultrasound and exhibited longer survival when treated with the combination of Folfirinox, ICB, and anti-SEMA4D antibody, compared to Folfirinox alone, Folfirinox plus ICB, or Folfirinox plus anti-SEMA4D antibody. Conclusions: Plexin B1+ myeloid subsets penetrate human PDAC tumors, and treatment with SEMA4D-blocking antibody improved response to ICB in combination with standard-of-care Folfirinox in preclinical murine studies. Future work will focus on understanding the immune mechanism of improved therapeutic response, as well as further characterization of the prevalence and prognostic ramifications of the SEMA4D-Plexin B axis in PDAC. Citation Format: Luis I. Ruffolo, Katherine M. Jackson, Nicholas Ullman, Alexander Chacon, Alexa Melucci, Paul Burchard, Mary Georger, Rachel Jewell, Peter Prieto, Brian Belt, Crystal Mallow, Elizabeth Evans, Terrence Fisher, Maurice Zauderer, Christina Wu, Brian Olson, Gregory B. Lesinski, David C. Linehan. Targeting the semaphorin 4D-plexin B axis to augment FOLFIRINOX in a murine model of pancreatic adenocarcinoma [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr B48.
- Published
- 2019
19. Abstract CT016: Integrated biomarker trials of VX15/2503 (pepinemab) in combination with checkpoint inhibitors in window of opportunity studies in solid tumors
- Author
-
Brian Olson, Michael C. Lowe, Maurice Zauderer, Desa Rae Pastore, Crystal Mallow, Conor E. Steuer, John E. Leonard, Ernest S. Smith, Elizabeth E. Evans, Gregory B. Lesinski, Nabil F. Saba, Christina Wu, Ragini R. Kudchadkar, and Terrence L. Fisher
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,business.industry ,T cell ,Melanoma ,Cancer ,Ipilimumab ,medicine.disease ,01 natural sciences ,010104 statistics & probability ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,Internal medicine ,medicine ,Biomarker (medicine) ,030212 general & internal medicine ,0101 mathematics ,Nivolumab ,business ,medicine.drug - Abstract
Interrogation of the tumor microenvironment (TME) is crucial to provide insight into biological activity, resistance mechanisms and implementation of rational combination immunotherapies. Semaphorin 4D (SEMA4D, CD100) has broad immunomodulatory effects in the TME. In preclinical models, antibody blockade of SEMA4D promoted immune infiltration and reduced function and recruitment of immunosuppressive myeloid cells within the TME. Importantly, preclinical combinations of anti-SEMA4D with immune checkpoint inhibitors (ICIs) enhanced T cell activity and tumor regression. VX15/2503 (pepinemab), an IgG4 humanized monoclonal antibody targeting SEMA4D, is currently being evaluated in window of opportunity, integrated biomarker trials to characterize immunomodulatory effects in pancreatic (PDAC), colorectal (CRC), and head and neck squamous cell (HNSCC) carcinomas, and melanoma. At present, three biomarker trials are recruiting patients with four resectable indications to investigate novel combinations of pepinemab with ICIs; 1) PDAC and CRC with resectable liver mets (NCT03373188, n=32), 2) HNSCC (NCT03690986, n=36), and 3) metastatic melanoma (NCT03769155, n=36). Prior to surgery, patients enroll in treatment cohorts including combinations of pepinemab with nivolumab and /or with ipilimumab, single agents, or no treatment. Three to seven weeks later, patients will undergo surgery and a substantial surgical section will be collected under the guidance of a pathologist for comparison across treatment groups and with a pre-dose tissue biopsy. Blood will be collected for PK, PD, and additional correlative biomarker assessments. The primary objective is to evaluate the treatment-induced effects on the immune profile in the TME and in peripheral blood. Additional objectives include, extending the previously reported safety profile of single agent pepinemab to ICI combination therapies, as well as exploring pathologic and radiographic responses in the melanoma study. Correlative multiplex flow cytometric flow panels have been established to phenotype cells in the TME and periphery. A multiplex IHC assay utilizing a sequential probe and strip procedure has also been qualified that allows co-localization, spatial orientation, and quantitation of multiple immune markers. Analysis of immune subsets include but are not limited to activated T cells, neutrophils, Treg cells, DCs, monocytes, macrophages, and importantly myeloid-derived suppressor cells (MDSCs). Target engagement and expression of SEMA4D and its cognate receptors will also be evaluated. Seven subjects have been enrolled in these studies as of 7 Jan 2019. These trials will provide the first integrated clinical assessment of anti-SEMA4D antibody activity to reprogram the TME. Citation Format: Gregory B. Lesinski, Terrence L. Fisher, Elizabeth E. Evans, John E. Leonard, Desa Rae Pastore, Crystal Mallow, Ernest Smith, Maurice Zauderer, Conor Steuer, Nabil F. Saba, Michael Lowe, Ragini R. Kudchadkar, Brian Olson, Christina Wu. Integrated biomarker trials of VX15/2503 (pepinemab) in combination with checkpoint inhibitors in window of opportunity studies in solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT016.
- Published
- 2019
20. Abstract 4074: Heat shock protein 90 inhibitors alter pancreatic stellate cell cytokine production and enhances the efficacy of immune checkpoint blockade in pancreatic cancer
- Author
-
Mohammad Zaidi, Yuchen Zhang, Michael B. Ware, Matthew R. Farren, Hannah Komar, Brian Olson, Ganji P. Nagaraju, Mehmet Akce, Olatunji Alese, Shishir Maithel, Juan Sarmiento, Walid Shaib, Christina Wu, Bassel El-Rayes, and Gregory B. Lesinski
- Subjects
Cancer Research ,Oncology - Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a prominent fibrotic stroma, which is a result of interactions between tumor, immune and pancreatic stellate cells (PSC). Prior work from our laboratory has defined a role for stroma-derived cytokines such as IL-6 as a significant barrier restraining immunity against PDAC. Our group is pursuing novel approaches to target pathways in the tumor microenvironment (TME), in an effort to improve access of effector immune cells to PDAC and response to immunotherapy. Heat shock protein-90 (Hsp90), is a chaperone protein and a versatile target in pancreatic cancer. Hsp90 regulates a diverse array of cellular processes of relevance to both the tumor and the immune system. However, to date the role of Hsp90 in PSC has not been explored in detail. We hypothesize that targeting Hsp90 can modulate the TME, through its ability to target inflammatory signaling and cytokine production by PSC and enhance the efficacy of immunotherapy. Treatment of immortalized and primary patient PSC with the Hsp90 inhibitor XL888 led to decreased IL-6 at the transcript and protein level in vitro. XL888 directly limited PSC growth, and reduced expression of alpha-SMA, Jak/STAT and MAPK signaling intermediates as determined via immunoblot. Combined therapy with XL888 and anti-PD-1 was efficacious in C57BL/6 mice bearing syngeneic subcutaneous (Panc02) or orthotopic (KPC-Luc) tumors, as compared to treatment with either agent alone. The treatment was well-tolerated, with no difference in body weight observed in either model. Laboratory studies are assessing immune and stromal biomarkers to define the impact on PSC, cytokines and T-cell biomarkers in the TME. Finally, we have completed the dose escalation phase of a Phase Ib/II clinical trial of XL888 (Hsp90i) and pembrolizumab (anti-PD-1) at our institution. Expansion cohorts of patients with metastatic pancreatic cancer (n=16) or colorectal cancer (n=16) are now accruing, with paired biopsy and peripheral blood samples to address the impact of Hsp90 inhibition on anti-PD-1 mediated T cell proliferation, cytokine production, and PSC-derived cytokine signatures. Citation Format: Mohammad Zaidi, Yuchen Zhang, Michael B. Ware, Matthew R. Farren, Hannah Komar, Brian Olson, Ganji P. Nagaraju, Mehmet Akce, Olatunji Alese, Shishir Maithel, Juan Sarmiento, Walid Shaib, Christina Wu, Bassel El-Rayes, Gregory B. Lesinski. Heat shock protein 90 inhibitors alter pancreatic stellate cell cytokine production and enhances the efficacy of immune checkpoint blockade in pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4074.
- Published
- 2019
21. Abstract 1545: Altered myeloid and lymphoid composition of tumor microenvironment following anti-SEMA4D and immune checkpoint combination therapies
- Author
-
Conor E. Steuer, Elizabeth E. Evans, Gregory B. Lesinski, Antoni Ribas, Maria Scrivens, Emily Greengard, Alan Howell, Brian Olson, Paul E. Clavijo, Ernest S. Smith, Siwen Hu-Lieskovan, Terrence L. Fisher, Clint T. Allen, Leslie Balch, Sebold Torno, Christina Wu, Christine Reilly, Maurice Zauderer, Nabil F. Saba, Crystal Mallow, Desa Rae Pastore, John E. Leonard, and Holm Bussler
- Subjects
Cancer Research ,Tumor microenvironment ,Myeloid ,LAG3 ,business.industry ,T cell ,Ipilimumab ,Immune checkpoint ,Avelumab ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Nivolumab ,business ,medicine.drug - Abstract
Anti-semaphorin 4D (SEMA4D, CD100) blocking antibody promotes immune infiltration, reduces immunosuppressive myeloid cells, and enhances T cell activity and tumor growth inhibition in combination with various immunotherapies in preclinical animal models. Clinical trials of immune checkpoint inhibitors (ICI) in combination with pepinemab (VX15/2503), humanized anti-SEMA4D antibody, are currently underway in several cancer indications. SEMA4D exerts multi-faceted effects within the tumor microenvironment by creating a barrier at the tumor-stroma margin to restrict immune cell infiltration and promote immunosuppressive activity of myeloid-derived cells. Blocking antibody to SEMA4D directly enhanced M1/M2 ratio and reduced both expression of chemokines that recruit MDSC and the ability of MDSC to suppress T cell activity. Antibody blockade simultaneously restored the ability of dendritic cells and cytotoxic T cells to infiltrate the TME, increasing ratio of Teffector to Tregulatory cells, in syngeneic tumor models. Importantly, anti-SEMA4D MAb enhanced the activity of co-administered immunotherapies, including antibodies to PD1, CTLA-4, and LAG3, epigenetic modulators, and additional novel immunomodulatory combinations in murine breast, colon, head and neck, and melanoma tumor models. New data will describe development and application of methods to assess immune phenotype and biomarkers in translational and clinical studies. These include flow cytometry and whole slide scans of multiplex IHC panels to examine MDSC, M1/M2 macrophage, monocytes, activated DC, B cells, exhausted, activated and stem-like populations of T cells in clinical samples. Expression of SEMA4D and its receptor PlexinB1 were also evaluated in spatial context and cellular co-localization. In summary, SEMA4D represents a novel target to regulate immune infiltration and mesenchymal suppression, sources of resistance to current immunotherapies. Pepinemab treatment was well tolerated in a Phase I oncology trial (NCT01313065) and is currently being evaluated as single agent or in ICI combinations in: (i) a Phase 1b/2a combination trial of pepinemab with avelumab in ICI naïve or ICI refractory NSCLC (CLASSICAL-Lung) (NCT03268057); (ii) a phase 1 combination trial of pepinemab with nivolumab or ipilimumab in melanoma patients who have progressed on any anti-PD-1/PD-L1 (NCT03425461); (iii) a neoadjuvant integrated biomarker trial in patients with metastatic colorectal, pancreatic (NCT03373188) and head and neck (NCT03690986) cancers treated with pepinemab in combination with nivolumab or ipilimumab; and (iv) a Phase 1/2 trial of pepinemab in children with solid tumors and children and young adults with osteosarcoma (NCT03320330). Clinical trials will evaluate safety, tolerability, efficacy, and biological endpoints, including immunophenotyping tumors and blood. Citation Format: Elizabeth E. Evans, Terrence L. Fisher, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Desa Rae Pastore, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Clint Allen, Paul E. Clavijo, Gregory Lesinski, Christina Wu, Conor Steuer, Nabil F. Saba, Brian Olson, Siwen Hu-Lieskovan, Antoni Ribas, Emily G. Greengard, Ernest S. Smith, Maurice Zauderer. Altered myeloid and lymphoid composition of tumor microenvironment following anti-SEMA4D and immune checkpoint combination therapies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1545.
- Published
- 2019
22. Abstract LB-235: COLOMATE: Colorectal cancer and liquid biopsy screening protocol for molecularly assigned therapy
- Author
-
Christina Wu, Pashtoon Murtaza Kasi, Daniel H. Ahn, Tanios Bekaii-Saab, Richard B. Lanman, John H. Strickler, Andrew B. Dodge, Kimmie Ng, Fang-Shu Ou, Kristen K. Ciombor, Tyler Zemla, Katrina S. Pedersen, Scott Kopetz, Rebecca Nagy, and Shumei Kato
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,Microsatellite instability ,Cancer ,medicine.disease ,Oxaliplatin ,Irinotecan ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Liquid biopsy ,Prospective cohort study ,business ,Allele frequency ,medicine.drug - Abstract
Background: Colorectal cancer (CRC) is the second leading cause of cancer death among men and women in the United States, and additional treatment strategies are needed for patients with advanced disease. Prospective therapeutic studies based on tissue-detected genomic targets such as ERBB2 (HER2), BRAF V600E, gene rearrangements/fusions, and microsatellite instability (MSI) have demonstrated clinical benefit for mCRC patients. Although correlative studies and/or retrospective case series utilizing circulating cell-free DNA (cfDNA) next generation sequencing (NGS) have shown that plasma detection of these genomic targets shows promise in mCRC, prospective studies are lacking. We hypothesize that comprehensive genomic profiling of cfDNA will identify actionable targets and improve outcomes for patients with mCRC. Methods: COLOMATE (Colorectal Cancer and Liquid Biopsy Screening Protocol for Molecularly Assigned Therapy; NCT03765736) is a phase II umbrella screening protocol sponsored by the Academic and Community Cancer Research United (ACCRU) consortium. This study utilizes Guardant360™ (Guardant Health, Redwood City, CA) - a blood-based targeted NGS panel - to identify actionable genomic alterations in mCRC patients and to assess the impact of molecularly assigned therapy accordingly under its individual companion studies. Eligible mCRC patients must have progressed on, been intolerant to, or have a contraindication to treatment with a fluoropyrimidine, oxaliplatin, irinotecan, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies (mAbs) and anti-epidermal growth factor receptor (EGFR) mAbs if RAS wild-type. Companion studies are dynamic and currently include those for patients whose tumors are either RAS/BRAF wild-type, HER2 amplified, FGFR altered, RAS mutated, or without any known actionable alteration. Additional companion studies are under active development. The primary objectives of this study are to perform blood-based genomic profiling on patients with treatment refractory mCRC to facilitate accrual to molecularly assigned therapies, and to obtain patient-matched tumor tissue and cell free DNA from peripheral blood to facilitate clinically annotated genomic analyses. Secondary correlative objectives are to explore mechanisms of acquired resistance to molecularly assigned therapy and to explore the correlation between cfDNA mutational burden (allele frequency, copy number) and clinical outcomes such as objective response rate, progression-free survival, and overall survival. Citation Format: Kristen K. Ciombor, Fang-Shu Ou, Andrew Dodge, Tyler Zemla, Christina Wu, Kimmie Ng, Katrina Pedersen, Shumei Kato, Pashtoon M. Kasi, Daniel Ahn, Rebecca Nagy, Richard Lanman, Scott Kopetz, John H. Strickler, Tanios Bekaii-Saab. COLOMATE: Colorectal cancer and liquid biopsy screening protocol for molecularly assigned therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-235.
- Published
- 2019
23. Abstract CT017: A Phase Ib of a combination of two chimeric (Trastuzumab-like and Pertuzumab-like) HER-2 B cell peptide vaccine emulsified in ISA 720 and nor-MDP adjuvant in patients with advanced solid tumors
- Author
-
Tanios Bekaii-Saab, Lai Wei, Robert Wesolowski, Daniel Ahn, Christina Wu, Maryam Lustberg, Amir Mortazavi, Bhuvaneswari Ramaswamy, Jay Overholser, and Pravin Kaumaya
- Subjects
Cancer Research ,Oncology - Abstract
Background: HER-2 is a transmembrane receptor that is overexpressed in multiple epithelial tumors. We have previously shown that a first generation HER-2 B-cell peptide vaccine targeting epitopes (628-647) and (316-339) was safe and relatively active. We recently developed a novel B-cell epitope specific vaccine (HER-Vaxx) consisting of epitopes derived from the extracellular domain of HER-2 that correspond to amino acid sequences 597-626 (pertuzumab binding site) and 266-296 (trastuzumab binding site). Methods: This first-in-human Phase 1b study evaluated the safety, optimal immunologic/biologic dose (OID/OBD) and immunogenicity of HER-Vaxx. Using a 3+3 dose escalation design, 3 patients (pts) were treated at each dose level (DL). If Results: 49 patients with metastatic and/or recurrent solid tumors and a median of 4 prior lines of chemotherapy received at least one inoculation of HER-vaxx. Common related toxicities include injection site reactions (56% grade 1/2 and 5% grade 3/4). There were no DLTs. The OID/OBD was found to be DL 2 (1.5 mg of each peptide vaccine).A total of 28 patients received ≥ 3 vaccinations including 2 with a partial response and 14 with stable disease. Of those, 6 patients received at least one 6-month boost. The vaccine generated sustained humoral response eliciting HER-2 specific antibodies in the majority of responding patients. Conclusion: We demonstrate that HER-Vaxx is well tolerated and able to generate sustained anti-HER-2 immune response. Given the promise, continuous development of the vaccine is ongoing at the suggested OBD in HER-2 overexpressing tumors. Citation Format: Tanios Bekaii-Saab, Lai Wei, Robert Wesolowski, Daniel Ahn, Christina Wu, Maryam Lustberg, Amir Mortazavi, Bhuvaneswari Ramaswamy, Jay Overholser, Pravin Kaumaya. A Phase Ib of a combination of two chimeric (Trastuzumab-like and Pertuzumab-like) HER-2 B cell peptide vaccine emulsified in ISA 720 and nor-MDP adjuvant in patients with advanced solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT017.
- Published
- 2019
24. Abstract B176: Sequential immunotherapy and association with clinical outcomes in advanced-stage cancer patients
- Author
-
Bassel F. El-Rayes, Walid L. Shaib, Bradley C. Carthon, Haydn T. Kissick, David H. Lawson, R. Donald Harvey, Mehmet Akce, Colleen Lewis, Viraj A. Master, Christina Wu, Yuan Liu, Mehmet Asim Bilen, Rathi N. Pillai, Julie M. Shabto, Ragini R. Kudchadkar, Hannah Collins, Suresh S. Ramalingam, Taofeek K. Owonikoko, Olatunji B. Alese, Dylan J. Martini, and Conor E. Steuer
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bladder cancer ,business.industry ,medicine.medical_treatment ,Immunology ,Head and neck cancer ,Cancer ,Phases of clinical research ,Immunotherapy ,medicine.disease ,Internal medicine ,medicine ,Prior Immunotherapy ,Lung cancer ,business ,Prospective cohort study - Abstract
Background: There are now six approved immune checkpoint inhibitors for several different malignancies including melanoma, head and neck cancer, lung cancer, and renal cell carcinoma. Given the increased number of available immunotherapeutic agents, more patients are presenting in clinic as candidates for sequential immunotherapy. However, the efficacy of sequential immunotherapy in a trial setting is unknown. We investigated the association between prior treatment with immune checkpoint inhibitors and clinical outcomes in patients treated with subsequent immunotherapy in a phase 1 clinical trial. Methods: We conducted a retrospective review of 90 advanced stage cancer patients treated on immunotherapy-based phase 1 clinical trials at Winship Cancer Institute between 2009 and 2017. We included 49 patients with an immune checkpoint-indicated histology (melanoma, lung cancer, head and neck cancer, and bladder cancer). Patients were then analyzed based on whether they had received at least one immune checkpoint inhibitor prior to enrollment. Overall survival (OS) and progression-free survival (PFS) were calculated in months from immunotherapy initiation on trial to date of death and clinical or radiographic progression, respectively. Clinical benefit (CB) was defined as a best response of complete response (CR), partial response (PR), or stable disease (SD). Univariate analysis (UVA) and multivariate analysis (MVA) were carried out using Cox proportional hazard or logistic regression model. Covariates included age, presence of liver metastases, number of prior lines of systemic therapy, histology, and Royal Marsden Hospital (RMH) risk group. Results: The median age was 67 years and most patients (78%) were men. The most common histologies were melanoma (61%) and lung/head and neck cancers (37%). The majority (81%) of patients were RMH good risk. More than half of patients (n=27, 55%) had received at least one immune checkpoint inhibitor prior to trial enrollment: ten received anti-PD-1, two received anti-CTLA-4, five received anti-PD-1/CTLA-4 combination therapy, and ten received multiple immune checkpoint inhibitors. In MVA, patients who had not received a prior immune checkpoint inhibitor had significantly longer OS (HR: 0.22, CI: 0.07-0.70, p=0.010). These patients also trended towards longer PFS (HR: 0.86, CI: 0.39-1.87, p=0.699) and higher chance of CB (HR: 2.52, CI: 0.49-12.97, p=0.268). Immunotherapy-naïve patients had substantially longer OS (24.3 vs 10.9 months) and PFS (5.1 vs. 2.8 months) than patients who had prior immunotherapy per Kaplan-Meier estimation. Conclusion: Optimal treatment options for oncology patients who progress on immune checkpoint inhibitors are lacking. In this study, patients who received at least one prior immune checkpoint inhibitor had worse clinical outcomes on immunotherapy-based phase 1 clinical trials than immune checkpoint-naïve patients. This suggests that further development of immunotherapy combination therapies is needed to improve clinical outcomes of these patients. The results from this study should be validated in a larger, prospective study. Citation Format: Mehmet Bilen, Dylan Martini, Yuan Liu, Colleen Lewis, Hannah Collins, Julie Shabto, Mehmet Akce, Haydn Kissick, Bradley Carthon, Walid Shaib, Olatunji Alese, Rathi Pillai, Conor Steuer, Christina Wu, David Lawson, Ragini Kudchadkar, Bassel El-Rayes, Viraj Master, Suresh Ramalingam, Taofeek Owonikoko, R. Donald Harvey. Sequential immunotherapy and association with clinical outcomes in advanced-stage cancer patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B176.
- Published
- 2019
25. Abstract PR10: Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination immunotherapy
- Author
-
Alan Howell, Emily Greengard, Holm Bussler, Paul E. Clavijo, Gregory Lesiniski, Terrence L. Fisher, Crystal Mallow, Christina Wu, Christine Reilly, Siwen Hu-Lieskovan, Ernest S. Smith, Antoni Ribas, Maurice Zauderer, Clint T. Allen, Leslie Balch, Maria Scrivens, Elizabeth E. Evans, John E. Leonard, and Sebold Torno
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunology ,Ipilimumab ,Immunotherapy ,Immune checkpoint ,Avelumab ,Cancer immunotherapy ,Blocking antibody ,medicine ,Cancer research ,Nivolumab ,business ,medicine.drug - Abstract
Purpose: Tumor growth inhibition by anti-semaphorin 4D (SEMA4D, CD100) blocking antibody is enhanced when combined with various immunotherapies in preclinical animal models. Immune checkpoint combinations with pepinemab (VX15/2503), a humanized anti-SEMA4D antibody, are currently being evaluated in several clinical trials. Methods: Expanded mechanistic studies in syngeneic preclinical models investigated the effect of SEMA4D blockade on immune contexture within the tumor microenvironment, as a single agent and in combination with various immunotherapy agents. Antitumor activity and immune response was characterized by immunohistochemistry, flow cytometry, functional assays, and cytokine, chemokine and gene expression analysis. Pepinemab (VX15/2503) is currently being evaluated as single agent or in combination with other immunotherapies in four clinical trials: (i) a phase 1b/2a combination trial of pepinemab with avelumab in NSCLC (CLASSICAL-Lung) (NCT03268057); (ii) a phase 1 combination trial of pepinemab with nivolumab or ipilimumab in melanoma patients who have progressed on any anti-PD-1/PD-L1 (NCT03373188); (iii) a neoadjuvant integrated biomarker trial in patients with metastatic colorectal and pancreatic cancers treated with pepinemab in combination with nivolumab or ipilimumab (NCT03373188); and (iv) a phase 1/2 trial of pepinemab in children with solid tumors and children and young adults with osteosarcoma (NCT03320330). Results: SEMA4D exerts multifaceted effects within the tumor microenvironment by creating a barrier at the tumor-stroma margin to restrict immune cell infiltration and promoting immunosuppressive activity of myeloid-derived cells. Blocking antibody to SEMA4D directly enhanced M1/M2 ratio and both reduced expression of chemokines that recruit MDSC and the ability of MDSC to suppress T-cell proliferation. Antibody blockade reduced the function of immunosuppressive myeloid and regulatory T-cells in the TME while simultaneously restoring the ability of dendritic cells and cytotoxic T-cells to migrate into the tumor in several syngeneic tumor models. Importantly, anti-SEMA4D MAb enhanced the activity of co-administered immunotherapies in murine colon, head and neck (HNSCC), and melanoma models. For example, anti-SEMA4D plus anti-CTLA-4 resulted in 100% survival and 90% complete tumor rejection (CR) (p Citation Format: Elizabeth E. Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Terrence L. Fisher, Clint Allen, Paul Clavijo, Gregory Lesiniski, Christina Wu, Siwen Hu-Lieskovan, Antoni Ribas, Emily Greengard, Ernest S. Smith, Maurice Zauderer. Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr PR10.
- Published
- 2019
26. Abstract 1762: Shifting the tumor microenvironment with first-in-class semaphorin 4D mab for combination immunotherapy
- Author
-
Holm Bussler, Emily Greengard, Elizabeth E. Evans, Maria Scrivens, Clint T. Allen, Leslie Balch, Alan Howell, Christina Wu, Ernest S. Smith, Antoni Ribas, Christine Reilly, Sebold Torno, Paul E. Clavijo, Siwen Hu-Lieskovan, Crystal Mallow, Terrence L. Fisher, Maurice Zauderer, Gregory B. Lesinski, and John E. Leonard
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,Melanoma ,Cancer ,Ipilimumab ,medicine.disease ,Immune checkpoint ,Avelumab ,Oncology ,Chemokine secretion ,Cancer research ,Medicine ,Nivolumab ,business ,medicine.drug - Abstract
Purpose: In preclinical models, tumor growth inhibition by anti-semaphorin 4D (SEMA4D, CD100) blocking antibody is enhanced when combined with various immunotherapies. Immune checkpoint combinations with humanized VX15/2503 anti-SEMA4D are currently being evaluated in several clinical trials. Methods: Expanded mechanistic studies in preclinical models investigate the effect of SEMA4D signaling through its Plexin receptors (PLXN) on MDSC function and chemokine secretion in the tumor microenvironment. Humanized VX15/2503 anti-SEMA4D is now also being evaluated as single agent or in combination with other immunotherapies in four clinical trials:: (i) a Phase 1b/2a combination trial of VX15/2503 with avelumab in NSCLC (CLASSICAL-Lung); (ii) a phase 1 combination trial with nivolumab or ipilimumab in melanoma patients who have progressed on any anti-PD-1/PD-L1; (iii) a neoadjuvant integrated biomarker trial in patients with metastatic colorectal and pancreatic cancers treated with VX15/2503 in combination with nivolumab or ipilimumab; and (iv) a Phase 1/2 trial of VX15/2503 in children with solid tumors and children and young adults with osteosarcoma. Results: SEMA4D plays a multi-faceted role within the tumor microenvironment by creating a barrier at the tumor-stroma margin to restrict immune cell infiltration and promoting immunosuppressive activity of myeloid-derived cells. SEMA4D directly enhanced ability of MDSC to suppress T cell proliferation and antibody blockade reversed these effects, both in vitro and in vivo. Furthermore, SEMA4D-PLXN signaling modulates expression of chemokines that recruit MDSC. Importantly, anti-SEMA4D MAb can enhance activity of co-administered immunotherapies in murine colon, head and neck (HNSCC), and melanoma models. For example, anti-SEMA4D plus anti-CTLA-4 results in 100% survival and 90% complete tumor rejection (CR) (p Citation Format: Elizabeth E. Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Terrence Fisher, Clint Allen, Paul E. Clavijo, Gregory Lesinski, Christina Wu, Siwen Hu-Lieskovan, Antoni Ribas, Emily G. Greengard, Ernest S. Smith, Maurice Zauderer. Shifting the tumor microenvironment with first-in-class semaphorin 4D mab for combination immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1762.
- Published
- 2018
27. Abstract 2607: Blood based biomarkers and association with clinical outcome (CO) in advanced stage patients (pts) treated with immunotherapy (IO)
- Author
-
Mehmet Asim Bilen, Bradley C. Carthon, Bassel F. El-Rayes, Haydn T. Kissick, Olatunji B. Alese, David H. Lawson, Rathi N. Pillai, Dylan J. Martini, Conor E. Steuer, Mehmet Akce, Viraj A. Master, Walid L. Shaib, Hannah Collins, R. Donald Harvey, Colleen Lewis, Christina Wu, Ragini R. Kudchadkar, Taofeek K. Owonikoko, Suresh S. Ramalingam, and Yuan Liu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Blood based biomarkers ,Proportional hazards model ,business.industry ,medicine.medical_treatment ,Advanced stage ,Cancer ,Immunotherapy ,Logistic regression ,medicine.disease ,Continuous variable ,Risk groups ,Internal medicine ,medicine ,business - Abstract
Background: Optimal biomarkers for cancer pts treated with IO are currently lacking. Markers of inflammation, such as neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR) are readily available and associated with poor outcomes. We investigated the association between these markers and CO in pts treated with IO. Methods: We conducted a retrospective review of 90 pts with advanced cancer treated on IO-based phase I trials at the Winship Cancer Institute of Emory University between 2009-2017. Baseline NLR, MLR, and PLR were treated as continuous variables and rescaled by their own standard deviation. Overall survival (OS) was measured from the first dose of IO to date of death or hospice referral. Progression-free survival (PFS) was determined from first dose of IO to clinical or radiographic progression or death. We defined clinical benefit (CB) as complete response, partial response, or stable disease. Univariate association (UVA) and multivariable analysis (MVA) were carried out using Cox proportional hazard model or logistic regression model. Baseline covariates included race, gender, ECOG PS, # of prior therapies, Royal Marsden Hospital (RMH) risk group, IO indication, and # of metastatic sites. Results: The median pt age was 63 years and most (59%) were men. The most common histologies were melanoma (33%) and GI cancers (22%). The majority (81%) were RMH good risk. 46% of pts had CB on IO. The median NLR, MLR, and PLR was 3.63, 0.48, and 182.65, respectively. Increased NLR, MLR, and PLR were all associated with worse OS, PFS, and chance of CB (Table 1). NLR, MLR and PLR are highly correlated to each other (Pearson correlation coefficients ≥ 0.8, all p < 0.0001). Conclusion: NLR, MLR, and PLR are strongly associated with CO in pts treated with IO. Prospective validation of these findings are warranted. Table 1: UVA and MVA of NLR, MRL, and PLR with CO OS PFSCBUVAMVAUVAMVAUVAMVAHR (CI)p-valueHR (CI)p-valueHR (CI)p-valueHR (CI)p-valueOR (CI)p-valueOR (CI)p-valueNLR1.37 (1.11-1.70)0.003*1.30 (1.02-1.66)0.031*1.42 (1.18-1.70) *statistically significant Citation Format: Dylan J. Martini, Yuan Liu, Colleen Lewis, Hannah Collins, Mehmet Akce, Haydn Kissick, Bradley C. Carthon, Walid L. Shaib, Olatunji B. Alese, Rathi Pillai, Conor E. Steuer, Christina Wu, David H. Lawson, Ragini Kudchadkar, Bassel El-Rayes, Viraj A. Master, Suresh Ramalingam, Taofeek K. Owonikoko, R Donald Harvey, Mehmet Asim Bilen. Blood based biomarkers and association with clinical outcome (CO) in advanced stage patients (pts) treated with immunotherapy (IO) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2607.
- Published
- 2018
28. Abstract CT088: A phase I trial combining MEK-1/2 inhibition in combination with 5-fluorouracil and radiation for locally-advanced rectal adenocarcinoma
- Author
-
Cynthia Timmers, Evan Wuthrick, Emily Chan, Amy Webb, Tanios Bekaii-Saab, Sameek Roychowdhury, Ryan Robb, Christina Wu, Lai Wei, and Terence M. Williams
- Subjects
0301 basic medicine ,Trametinib ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Total mesorectal excision ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,Fluorouracil ,Internal medicine ,medicine ,Rectal Adenocarcinoma ,030211 gastroenterology & hepatology ,business ,medicine.drug - Abstract
Introduction: 5-fluorouracil (5-FU)-based chemoradiation (CRT) is a standard neoadjuvant treatment strategy for the treatment of locally-advanced rectal adenocarcinomas (LARC). MEK-1/2 inhibition has been shown to radiosensitize Ras mutant tumors in preclinical models, including colorectal cancer. In this phase 1 trial, we are testing MEK-1/2 inhibition in combination with neoadjuvant CRT for LARC. Methods: This trial is a standard 3+3 design testing 3 dose levels of the MEK-1/2 inhibitor trametinib in combination with CRT: 0.5 mg, 1.0 mg, and 2.0 mg daily, followed by an expansion cohort. All patients first receive a one week lead-in dosing of trametinib monotherapy, and undergo a tumor biopsy at the end of that week to assess pharmacodynamic inhibition by immunohistochemistry (IHC). Then, patients receive standard CRT (5-FU 225 mg/m2 per day by continuous infusion during radiotherapy, and 50.4 Gy in 28 fractions, 5 days/ week) in combination with trametinib (5 days/week). Patients have total mesorectal excision (TME) 6-10 weeks after completion of CRT. Results: 17 patients have been enrolled to date, with current enrollment occurring on the trametinib 2 mg expansion cohort (total 18 patients planned). Of these, 15 patients (10 males, 5 females) have toxicity and surgical response data available. Median age is 53 years (range 35-74 years), and 6 patients have Stage II disease, while 9 patients with Stage III disease. There have been no grade 4-5 toxicities. One dose-limiting toxicity (DLT) of diarrhea occurred in the 2mg dose cohort, attributed mainly to 5FU CRT, but led to trametinib discontinuation. Generalized skin rash led to trametinib discontinuation after 2 weeks of therapy for 1 patient and was held for 3 days for 2 patients. All patients completed TME, and 3 patients had wound infection at 2 weeks after TME, while 2 patients had perineal wound infection at 4 weeks after TME. Three out of 9 (33%) patients at the 2mg dose cohort had a pathologic complete response (pCR), and 2/9 (22%) patients had a near pCR. Adjuvant chemotherapy was given to 10 patients, 4 patients did not receive (3 refused), and 1 patient is pending. Correlative research studies have been performed in the first 12 patients enrolled. Examination of phospho-ERK levels using Vectra quantitative immunohistochemistry in pre-treatment versus post-lead-in trametinib tumor tissue reveals dose-dependent decreases in p-ERK expression in both tumor and stromal tissue as trametinib is increased from 0.5 to 2.0 mg (range 18-46% absolute reduction), with maximal reduction observed in the 2 mg cohort. Additionally, we have performed whole exome sequencing of a custom panel of 279 cancer associated genes (IGNITE platform) in tumor and normal tissue, and have identified high frequency mutations in KRAS, BRAF, and TP53, as well as additional lower frequency mutations in every tumor. One patient was noted to have microsatellite instability. Finally, we will present data correlating degree of change in p-ERK staining, mutational analysis, and mismatch repair status with pathologic response. Conclusions: The combination of trametinib with 5-FU-based CRT for LARC is feasible with promising signs of clinical activity and tolerability. Initial analysis of tumor tissue confirms dose-dependent pharmacodynamic inhibition of ERK-1/2 within 5 days of starting trametinib. Citation Format: Terence M. Williams, Christina Wu, Lai Wei, Emily Chan, Ryan Robb, Sameek Roychowdhury, Amy Webb, Cynthia Timmers, Tanios Bekaii-Saab, Evan Wuthrick. A phase I trial combining MEK-1/2 inhibition in combination with 5-fluorouracil and radiation for locally-advanced rectal adenocarcinoma [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 CT088. doi:10.1158/1538-7445.AM2017-CT088
- Published
- 2017
29. Abstract CT025: A phase I trial combining MEK-1/2 inhibition in combination with 5-fluorouracil and radiation for KRAS, BRAF, and NRAS mutant locally advanced rectal adenocarcinoma
- Author
-
Terence M. Williams, Tanios Bekaii-Saab, Cynthia Timmers, Evan Wuthrick, Ryan Robb, and Christina Wu
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Trametinib ,Oncology ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,medicine.disease_cause ,Surgery ,Tolerability ,Internal medicine ,medicine ,KRAS ,business ,Neoadjuvant therapy ,V600E - Abstract
Introduction: 5-fluorouracil (5-FU)-based chemoradiation (CRT) is a standard neoadjuvant treatment strategy for the treatment of locally-advanced rectal adenocarcinomas (LARC). MEK-1/2 inhibition has been shown to radiosensitize Ras mutant tumors in preclinical models, including colorectal cancer. In this phase 1 trial, we will test MEK-1/2 inhibition in combination with neoadjuvant CRT for LARC. Methods: This trial is a standard 3+3 design testing 3 dose levels of the MEK-1/2 inhibitor trametinib (GSK1120212) in combination with CRT: 0.5 mg, 1.0 mg, and 2.0 mg daily, followed by an expansion cohort. All patients first receive a one week lead-in dosing of trametinib monotherapy, and undergo a tumor biopsy at the end of that week to assess pharmacodynamic inhibition by immunohistochemistry (IHC). Then, patients receive standard CRT (5-FU 225 mg/m2 per day by continuous infusion during radiotherapy, and 50.4 Gy in 28 fractions, 5 days/ week) in combination with trametinib (5 days/week). Patients have total mesorectal excision (TME) 6-10 weeks after completion of CRT. Results: The trial opened in 2013, and 31 patients have been screened for KRAS/BRAF/NRAS activating mutations, of which 11 (35.4%) tested positive for a mutation. Of these, 9 patients (8 male, 1 female) have been enrolled so far, with 3 patients accrued in each dose level. Regarding mutation status, six patients have KRAS mutations (five G12V, one G13D), two patients have BRAF mutations (one V600E, one D594N), and one patient is unknown (in dose level 1). Age of patients ranges from 38-64 years old (median 55). All patients have successfully completed neoadjuvant therapy and TME (5 low anterior resection, 4 abdominal-perineal resection). No dose-limiting toxicities were observed on dose levels 1 and 2, and additional patients are currently being accrued onto dose level 3 (2.0 mg). One patient in dose level 3 had pathologic complete response (pCR) at the time of TME. Examination of pre-treatment tumor tissue by IHC reveals baseline hyperactivation of ERK-1/2 (p42/44 MAPK) in most tumors with some heterogeneity in levels of phospho-ERK staining, reflecting hyperactivation of the RAS/RAF-MAPK pathway. Qualitatively, ERK-1/2 activity is reduced from pre-treatment to post-trametinib lead-in tumor tissue, particularly in patients on dose level 3. The patient who experienced a pCR had the most significant reduction in ERK-1/2 activity in tumor tissue and tumor-associated stroma. We will also present quantitative data demonstrating ERK-1/2 inhibition using Vectra quantitative immunohistochemistry. Conclusions: The combination of trametinib with 5-FU-based CRT for LARC is feasible with promising signs of clinical activity and tolerability. Ongoing enrollment will further assess safety, tolerability, and determine the maximally-tolerated dose of trametinib. Initial analysis of tumor tissue suggests dose-dependent pharmacodynamic inhibition of ERK-1/2 within 5 days of starting trametinib. Citation Format: Terence M. Williams, Evan Wuthrick, Christina Wu, Ryan Robb, Cynthia Timmers, Tanios Bekaii-Saab. A phase I trial combining MEK-1/2 inhibition in combination with 5-fluorouracil and radiation for KRAS, BRAF, and NRAS mutant locally advanced rectal adenocarcinoma. [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 CT025.
- Published
- 2016
30. Abstract 5424: Therapeutic targeting of KRAS mutation-driven tumor progression in colorectal cancer
- Author
-
Ching-Shih Chen, Tanios Bekaii-Saab, Hsiao-Ching Chuang, Peng Chan Lin, Christina Wu, Po-Chen Chu, and Samuel K. Kulp
- Subjects
Cancer Research ,Colorectal cancer ,business.industry ,MEK inhibitor ,Cancer ,medicine.disease ,medicine.disease_cause ,Metastasis ,Oncology ,Growth factor receptor ,Tumor progression ,medicine ,Cancer research ,KRAS ,business ,Protein kinase B - Abstract
KRAS is the most frequently altered gene in colorectal cancer (CRC), with mutations occurring in 30-40% of colorectal cancer. Although KRAS mutation is associated with poor prognosis and resistance to anti-epidermal growth factor receptor therapy, the mechanism by which it promotes tumor metastasis remains undefined. Our study explored a new hypothesis that targeting the Y-box binding protein-1 (YB-1)-insulin-like growth factor-I receptor (IGF-IR) signaling axis which is downstream of KRAS, with a novel integrin-linked kinase inhibitor, T315, represents a therapeutically relevant strategy to block KRAS mutation-driven tumor progression. In liver metastases from CRC patients, we observed that there was a preponderance (79/108) of over-expression in both YB-1 and IGF-1R by immunohistochemistry. In colon cancer cell lines HCT-116 and SW480, knockdown and over-expression of KRAS affected the protein and mRNA levels of IGF-1R in a dose-dependent manner. But KRAS only affected the protein level and not mRNA of YB-1, thus suggesting that KRAS regulated YB-1 expression at the post-transcriptional level. Manipulation of YB-1 via plasmid overexpression and siRNA affected IGF-1R protein and mRNA levels in a dose-dependent manner, and YB-1 and IGF-1R promoter binding was confirmed via ChIP assay. There was a dose-dependent decrease in YB-1 and IGF-1R protein levels with MEK162, a MEK inhibitor, but not with LY294002m a PI3K inhibitor. This demonstrated that the KRAS regulation of the YB-1- IGFR-1R signaling axis to be via the MEK signaling pathway and not PI3K/Akt. Our novel drug T315 targets YB-1, and was shown to inhibit cell viability and cell migration in a dose-dependent manner. T315 treatment of the colon cancer cells also decreased protein levels of YB-1 and IGF-1R and affected epidermal mesenchymal transition markers such as E-cadherin, vimentin, and snail. These results suggest that a combination of a MEK inhibitor and T315 may be an effective therapeutic strategy to target KRAS mutation driven CRC. Citation Format: Christina Wu, Peng-Chan Lin, Po-Chen Chu, Hsiao-Ching Chuang, Samuel Kulp, Tanios Bekaii-Saab, Ching-Shih Chen. Therapeutic targeting of KRAS mutation-driven tumor progression in colorectal cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5424. doi:10.1158/1538-7445.AM2015-5424
- Published
- 2015
31. Abstract A76: Enhancement of cetuximab-induced antibody-dependent cellular cytotoxicity with lenalidomide in advanced solid tumors: A Phase I trial
- Author
-
Gregory A. Otterson, Jeffrey S. Rose, Christina Wu, William E. Carson, Tanios Bekaii-Saab, Panayiotis S. Savvides, Michael R. Grever, Miguel Villalona, Richard M. Goldberg, Erin M. Bertino, and Gerard Lozanski
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Cetuximab ,business.industry ,T cell ,Cancer ,Pharmacology ,medicine.disease_cause ,medicine.disease ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Panitumumab ,Interferon gamma ,KRAS ,business ,Lenalidomide ,medicine.drug - Abstract
Introduction: Antibody-dependent cellular cytotoxicity (ADCC) is one mechanism of action of monoclonal antibody (mAb) therapy. ADCC occurs via the innate immune system's ability to recognize mAb coated cancer cells and activate effector cells. Lenalidomide is an immunomodulatory agent with capacity to stimulate T cell proliferation, activate natural killer cells, and effect immune cytokines including IL-2, IL-12, and interferon gamma. Both preclinical and clinical data demonstrate the ability of lenalidomide to increase ADCC activity of mAb therapy. This Phase I CTEP sponsored trial aims to evaluate the combination of cetuximab with lenalidomide to enhance ADCC activity in advanced colorectal (CRC) and head and neck squamous cell cancers (HNSCC). Patients/Methods: This Phase I dose escalation trial included patients with locally advanced or metastatic CRC (KRAS wild type) or HNSCC. Treatment consisted of cetuximab 500 mg/m2 IV every 2 weeks with lenalidomide orally days 1-21 every 28 days. Cetuximab dose was the same in all cohorts although dose reductions were permitted for toxicity. Three dose levels of lenalidomide were evaluated (15 mg, 20 mg, and 25 mg). Prior treatment with cetuximab, panitumumab, or other EGFR directed therapy was allowed. Planned correlative studies include measurement of ADCC using an in vitro chromium release assay, serum interferon gamma and NK cytokine profiles, and Fc gamma receptor polymorphisms. Results: A total of 22 patients have been treated (19 CRC, 3 HNSCC). Grade 3 fatigue has been the only observed dose-limiting toxicity. One partial response was observed and 7 patients had stable disease as best response. Two patients remain on treatment. The recommended Phase II dose is cetuximab 500 mg/m2 with lenalidomide 25 mg daily days 1-21. Conclusions: Cetuximab and lenalidomide was well-tolerated with minimal activity. Although response was not a primary endpoint, there is evidence of anti-tumor activity and clinical efficacy. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A76. Citation Format: Erin M. Bertino, Jeffrey S. Rose, Christina Wu, Tanios Bekaii-Saab, Panayiotis S. Savvides, Richard M. Goldberg, Miguel Villalona, Gerard Lozanski, William Carson, Michael Grever, Gregory Otterson. Enhancement of cetuximab-induced antibody-dependent cellular cytotoxicity with lenalidomide in advanced solid tumors: A Phase I trial. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A76.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.