Try a new search

Format these results:

Searched for:

in-biosketch:true

person:obersp01

Total Results:

73


S2303: phase II/III trial of paclitaxel + ramucirumab ± nivolumab in gastric and esophageal adenocarcinoma (PARAMUNE)

Saeed, Anwaar; Colby, Sarah; Oberstein, Paul Eliezer; Duda, Dan G; Park, Robin; Agarwal, Rajiv; Figueroa-Moseley, Colmar; Vaidya, Riha; Unger, Joseph M; Guthrie, Katherine A; Rocha, Flavio G; Senthil, Maheswari; Safyan, Rachael A; Wainberg, Zev A; Iqbal, Syma; Chiorean, E Gabriela; Philip, Philip A
NCT06203600.
PMID: 40155326
ISSN: 1744-8301
CID: 5817862

Association of tumor microbiome with survival in resected early-stage PDAC

Meng, Yixuan; Wang, Chan; Usyk, Mykhaylo; Kwak, Soyoung; Peng, Chengwei; Hu, Kenneth S; Oberstein, Paul E; Krogsgaard, Michelle; Li, Huilin; Hayes, Richard B; Ahn, Jiyoung
The pancreas tumor microbiota may influence tumor microenvironment and influence survival in early-stage pancreatic ductal adenocarcinoma (PDAC); however, current studies are limited and small. We investigated the relationship of tumor microbiota to survival in 201 surgically resected patients with localized PDAC (Stages I-II), from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts. We characterized the tumor microbiome using RNA-sequencing data. We examined the association of the tumor microbiome with overall survival (OS), via meta-analysis with the Cox PH model. A microbial risk score (MRS) was calculated from the OS-associated microbiota. We further explored whether the OS-associated microbiota is related to host tumor immune infiltration. PDAC tumor microbiome α- and β-diversities were not associated with OS; however, 11 bacterial species, including species of Gammaproteobacteria, confirmed by extensive resampling, were significantly associated with OS (all Q < 0.05). The MRS summarizing these bacteria was related to a threefold change in OS (hazard ratio = 2.96 per standard deviation change in the MRS, 95% confidence interval = 2.26-3.86). This result was consistent across the two cohorts and in stratified analyses by adjuvant therapy (chemotherapy/radiation). Identified microbiota and the MRS also exhibited association with memory B cells and naïve CD4+ T cells, which may be related to the immune landscape through BCR and TCR signaling pathways. Our study shows that a unique tumor microbiome structure, potentially affecting the tumor immune microenvironment, is associated with poorer survival in resected early-stage PDAC. These findings suggest that microbial mechanisms may be involved in PDAC survival, potentially informing PDAC prognosis and guiding personalized treatment strategies.IMPORTANCEMuch of the available data on the PDAC tumor microbiome and survival are derived from relatively small and heterogeneous studies, including those involving patients with advanced stages of pancreatic cancer. There is a critical knowledge gap in terms of the tumor microbiome and survival in early-stage patients treated by surgical resection; we expect that advancements in survival may initially be best achieved in these patients who are treated with curative intent.
PMID: 40013793
ISSN: 2379-5077
CID: 5801172

A Phase 2 Study of Sotigalimab, a CD40 Agonist Antibody, Plus Concurrent Chemoradiation as Neoadjuvant Therapy for Esophageal and Gastroesophageal Junction Cancers

Ko, Andrew H; Chao, Joseph; Noel, Marcus S; Shankaran, Veena; Sohal, Davendra; Crow, Mary; Oberstein, Paul E; Scott, Aaron J; McRee, Autumn J; Rocha Lima, Caio Max Sao Pedro; Fong, Lawrence; Keenan, Bridget P; Soto, Maira; Filbert, Erin L; Hsu, Frank J; Yang, Xiaodong
PURPOSE/OBJECTIVE:Neoadjuvant chemoradiation (NCRT) followed by surgical resection represents a standard approach for patients with locally advanced esophageal/GEJ cancers. Sotigalimab is a high affinity CD40 agonist antibody capable of inducing and expanding anti-tumor immune responses by activating dendritic cells, T and B lymphocytes, NK cells, and M1 macrophages. This study examined the safety and efficacy of combining sotigalimab with NCRT in patients with esophageal or GEJ cancers. PATIENTS AND METHODS/METHODS:Patients with resectable (T1-3, Nx) adenocarcinoma(AC) or squamous cell carcinoma(SCC) of the esophagus or GEJ were eligible. T1N0 and cervical tumors were excluded. Study treatment: weekly carboplatin/paclitaxel with concurrent radiation 5040 cGy plus 3-4 doses of sotigalimab prior to Ivor-Lewis esophagectomy. Primary efficacy endpoint was pathologic complete response (path CR) rate. RESULTS:33 patients were enrolled (AC 76%, SCC 24%; clinical stage III 67%). Ninety percent of patients received all planned doses of sotigalimab. The most common adverse events attributed to sotigalimab were nausea, fever/chills, fatigue, and cytokine release syndrome (CRS); most of these were Grade 1-2. Grade >3 CRS was observed in 3 pts (9%). Twenty-five of the 29 efficacy-evaluable patients underwent an R0 resection (87.9%), with an overall path CR rate of 37.9% (11/29). Post-tumor samples demonstrated increased infiltration and activation of dendritic cells, monocytes, and cytotoxic T cells compared to baseline. CONCLUSIONS:Sotigalimab combined with NCRT for esophageal or GEJ cancers was generally well tolerated and achieved path CR rates that compare favorably to historical data and are promising for this treatment strategy. CLINICAL TRIAL INFORMATION/BACKGROUND:NCT03165994.
PMID: 39907035
ISSN: 2767-9764
CID: 5783922

Relacorilant plus nab-paclitaxel for the treatment of metastatic pancreatic ductal adenocarcinoma: results from the open-label RELIANT study

Borazanci, Erkut H; Bahary, Nathan; Chung, Vincent; Huyck, Timothy K; Kio, Ebenezer A; Chiorean, Elena Gabriela; Skeel, Roland T; Alese, Olatunji B; Cardin, Dana B; Fountzilas, Christos; Hanna, Wahid T; Leal, Alexis D; Lee, Valerie; Noonan, Anne M; Philip, Philip A; Wainberg, Zev A; Pashova, Hristina; Mann, Grace; Oberstein, Paul E
BACKGROUND:Modulation of glucocorticoid receptor (GR) activity in tumor cells enhances chemotherapy efficacy. We evaluated the selective GR modulator relacorilant plus nab-paclitaxel in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who had received at least 2 prior therapy lines. PATIENTS AND METHODS/METHODS:In this open-label, single-arm, phase III study, patients received once-daily oral relacorilant (100 mg, titrated to 150 mg in 25 mg increments/cycle) and nab-paclitaxel (80 mg/m2) on days 1, 8, and 15 of 28-day cycles. The primary efficacy endpoint was objective response rate (ORR) by blinded independent central review. Progression-free survival (PFS), overall survival (OS), target gene modulation, and safety were also assessed. RESULTS:Of 43 patients enrolled, 31 were evaluable for ORR (12 did not reach first postbaseline radiographic assessment). An interim analysis to assess whether ORR was ≥10% showed no confirmed responses and the study was discontinued. Two (6.5%) patients attained unconfirmed partial responses and 15 (48.4%) had stable disease. Fourteen of 31 (45.2%) patients had reductions in target lesion size, despite prior nab-paclitaxel exposure in 12 of the 14. Median PFS and OS were 2.4 months (95% CI, 1.4-4.2) and 3.9 months (95% CI, 2.8-4.9), respectively. The most common adverse events were fatigue and nausea. RNA analysis confirmed that relacorilant plus nab-paclitaxel suppressed 8 cortisol target genes of interest. CONCLUSION/CONCLUSIONS:Relacorilant plus nab-paclitaxel showed modest antitumor activity in heavily pretreated patients with mPDAC, with no new safety signals. Studies of this combination in other indications with a high unmet medical need are ongoing.
PMID: 39191530
ISSN: 1549-490x
CID: 5729692

Impact of Adjuvant Chemotherapy on Resected Intraductal Papillary Mucinous Neoplasm-Derived Pancreatic Cancer: Results From an International Multicenter Study

Habib, Joseph R; Kinny-Köster, Benedict; Javed, Ammar A; Zelga, Poitr; Saadat, Lily V; Kim, Rachel C; Gorris, Myrte; Allegrini, Valentina; Watanabe, Shuichi; Sharib, Jeremy; Arcerito, Massimo; Kaiser, Jörg; Lafaro, Kelly J; Tu, Min; Bhandre, Manish; Shi, Chanjuan; Kim, Michael P; Correa, Camilo; Daamen, Lois A; Oberstein, Paul E; Schmidt, C Max; Hanna, Nader N; Allen, Peter; Loos, Martin; Shrikhande, Shailesh V; Molenaar, I Quintus; Frigerio, Isabella; Katz, Matthew H G; Soares, Kevin C; Miao, Yi; Del Chiaro, Marco; He, Jin; Hackert, Thilo; Salvia, Roberto; Büchler, Markus W; Castillo, Carlos Fernandez-Del; Besselink, Marc G; Marchegiani, Giovanni; Wolfgang, Christopher L; ,
PURPOSE/OBJECTIVE:The benefit of adjuvant therapy for intraductal papillary mucinous neoplasm (IPMN)-derived pancreatic ductal adenocarcinoma (PDAC) remains unclear because of severely limited evidence. Although biologically distinct entities, adjuvant therapy practices for IPMN-derived PDAC are largely founded on pancreatic intraepithelial neoplasia-derived PDAC. We aimed to evaluate the role of adjuvant chemotherapy in IPMN-derived PDAC. METHODS:This international multicenter retrospective cohort study (2005-2018) was conceived at the Verona Evidence-Based Medicine meeting. Cox regressions were performed to identify risk-adjusted hazard ratios (HR) associated with overall survival (OS). Kaplan-Meier curves and log-rank tests were employed for survival analysis. Logistic regression was performed to identify factors motivating adjuvant chemotherapy administration. A decision tree was proposed and categorized patients into overtreated, undertreated, and optimally treated cohorts. RESULTS:> .05). Based on this model, we observed undertreatment in 18.1% and overtreatment in 61.2% of patients. Factors associated with chemotherapy administration included younger age, R1-margin, poorer differentiation, and nodal disease. CONCLUSION/CONCLUSIONS:Almost half of patients with resected IPMN-derived PDAC may be overtreated or undertreated. In patients with node-negative disease or normal CA19-9, adjuvant chemotherapy is not associated with a survival benefit, whereas those with node-positive disease and elevated CA19-9 have an associated benefit from adjuvant chemotherapy. A decision tree was proposed. Randomized controlled trials are needed for validation.
PMID: 39255450
ISSN: 1527-7755
CID: 5690222

Omission of 5-Fluorouracil Bolus From Multidrug Regimens for Advanced Gastrointestinal Cancers: A Multicenter Cohort Study

Peng, Chengwei; Saffo, Saad; Oberstein, Paul E; Shusterman, Michael; Thomas, Charlene; Becker, Daniel J; Berlin, Jordan D; Leichman, Lawrence P; Boursi, Ben; Nagar, Anil B; Yu, Shun
BACKGROUND:5-Fluorouracil (5-FU) is a major component of gastrointestinal cancer treatments. In multidrug regimens such as FOLFOX, FOLFIRI, and FOLFIRINOX, 5-FU is commonly administered as a bolus followed by an infusion. However, the pharmacologic rationale for incorporating the 5-FU bolus in these regimens is unclear, and there are other effective regimens for gastrointestinal cancers that do not include the bolus. The purpose of this study was to determine whether omission of the 5-FU bolus was associated with a difference in survival and toxicity. METHODS:A real-world database from Flatiron Health was queried for patients with advanced colorectal, gastroesophageal, and pancreatic cancers who received first-line FOLFOX, FOLFIRI, and FOLFIRINOX regimens. Cox proportional hazards and Kaplan-Meier analyses were performed to compare survival outcomes between patients who received the 5-FU bolus and those who did not. Inverse probability of treatment weighted (IPTW) analysis was performed to adjust for treatment selection bias. RESULTS:This study included 11,765 patients with advanced colorectal (n=8,670), gastroesophageal (n=1,481), and pancreatic (n=1,614) cancers. Among all first-line 5-FU multidrug regimens, 10,148 (86.3%) patients received a 5-FU bolus and 1,617 (13.7%) did not. After IPTW analysis, we found that omitting the bolus was not associated with a decrease in overall survival (hazard ratio, 0.99; 95% CI, 0.91-1.07; P=.74). However, omitting the bolus was associated with reductions in neutropenia (10.7% vs 22.7%; P<.01), thrombocytopenia (11.2% vs 16.1%; P<.01), and use of granulocyte colony-stimulating factors after treatment (19.6% vs 29.1%; P<.01). CONCLUSIONS:After adjusting for baseline clinical factors, we found that omission of the 5-FU bolus from FOLFOX, FOLFIRI, and FOLFIRINOX regimens was not associated with decreased survival, but resulted in decreased toxicity and possible health care savings.
PMID: 39236754
ISSN: 1540-1413
CID: 5806132

Blockade of IL1β and PD1 with Combination Chemotherapy Reduces Systemic Myeloid Suppression in Metastatic Pancreatic Cancer with Heterogeneous Effects in the Tumor

Oberstein, Paul E; Dias Costa, Andressa; Kawaler, Emily A; Cardot-Ruffino, Victoire; Rahma, Osama E; Beri, Nina; Singh, Harshabad; Abrams, Thomas A; Biller, Leah H; Cleary, James M; Enzinger, Peter; Huffman, Brandon M; McCleary, Nadine J; Perez, Kimberly J; Rubinson, Douglas A; Schlechter, Benjamin L; Surana, Rishi; Yurgelun, Matthew B; Wang, S Jennifer; Remland, Joshua; Brais, Lauren K; Bollenrucher, Naima; Chang, Eugena; Ali, Lestat R; Lenehan, Patrick J; Dolgalev, Igor; Werba, Gregor; Lima, Cibelle; Keheler, C Elizabeth; Sullivan, Keri M; Dougan, Michael; Hajdu, Cristina; Dajee, Maya; Pelletier, Marc R; Nazeer, Saloney; Squires, Matthew; Bar-Sagi, Dafna; Wolpin, Brian M; Nowak, Jonathan A; Simeone, Diane M; Dougan, Stephanie K
Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1β blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1β (IL1β), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1β blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.
PMCID:11369625
PMID: 38990554
ISSN: 2326-6074
CID: 5687222

Somatic Mutational Analysis in EUS-Guided Biopsy of Pancreatic Adenocarcinoma: Assessing Yield and Impact

Dong, Sue; Agarunov, Emil; Fasullo, Matthew; Kim, Ki-Yoon; Khanna, Lauren; Haber, Gregory; Janec, Eileen; Simeone, Diane; Oberstein, Paul; Gonda, Tamas
OBJECTIVES/OBJECTIVE:We sought to determine the yield of somatic mutational analysis from EUS-guided biopsies of pancreatic adenocarcinoma compared to that of surgical resection and to assess the impact of these results on oncologic treatment. METHODS:We determined the yield of EUS sampling and surgical resection. We evaluated the potential impact of mutational analysis by identifying actionable mutations and its direct impact by reviewing actual treatment decisions. RESULTS:Yield of EUS sampling was 89.5%, comparable to the 95.8% yield of surgical resection. Over a quarter in the EUS cohort carried actionable mutations, and of these, over one in six had treatment impacted by mutational analysis. CONCLUSIONS:EUS sampling is nearly always adequate for somatic testing and may have substantial potential and real impact on treatment decisions.
PMID: 38546128
ISSN: 1572-0241
CID: 5645102

Devimistat (CPI-613) With Modified Fluorouarcil, Oxaliplatin, Irinotecan, and Leucovorin (FFX) Versus FFX for Patients With Metastatic Adenocarcinoma of the Pancreas: The Phase III AVENGER 500 Study

Philip, Philip A; Sahai, Vaibhav; Bahary, Nathan; Mahipal, Amit; Kasi, Anup; Rocha Lima, Caio Max S; Alistar, Angela T; Oberstein, Paul E; Golan, Talia; Metges, Jean-Philippe; Lacy, Jill; Fountzilas, Christos; Lopez, Charles D; Ducreux, Michel; Hammel, Pascal; Salem, Mohamed; Bajor, David; Benson, Al B; Luther, Sanjeev; Pardee, Timothy; Van Cutsem, Eric
PURPOSE/OBJECTIVE:Metastatic pancreatic adenocarcinoma (mPC) remains a difficult-to-treat disease. Fluorouarcil, oxaliplatin, irinotecan, and leucovorin (FFX) is a standard first-line therapy for mPC for patients with a favorable performance status and good organ function. In a phase I study, devimistat (CPI-613) in combination with modified FFX (mFFX) was deemed safe and exhibited promising efficacy in mPC. METHODS:total per day on days 1 and 3 in the experimental arm. The primary end point of the study was overall survival (OS). RESULTS:11.5%), respectively. CONCLUSION/CONCLUSIONS:Devimistat in combination with mFFX did not improve long- and short-term mPC patient outcomes compared with standard FFX. There were no new toxicity signals with the addition of devimistat.
PMID: 39088774
ISSN: 1527-7755
CID: 5696562

Altered salivary microbiota associated with high-sugar beverage consumption

Fan, Xiaozhou; Monson, Kelsey R; Peters, Brandilyn A; Whittington, Jennifer M; Um, Caroline Y; Oberstein, Paul E; McCullough, Marjorie L; Freedman, Neal D; Huang, Wen-Yi; Ahn, Jiyoung; Hayes, Richard B
The human oral microbiome may alter oral and systemic disease risk. Consuming high sugar content beverages (HSB) can lead to caries development by altering the microbial composition in dental plaque, but little is known regarding HSB-specific oral microbial alterations. Therefore, we conducted a large, population-based study to examine associations of HSB intake with oral microbiome diversity and composition. Using mouthwash samples of 989 individuals in two nationwide U.S. cohorts, bacterial 16S rRNA genes were amplified, sequenced, and assigned to bacterial taxa. HSB intake was quantified from food frequency questionnaires as low (< 1 serving/week), medium (1-3 servings/week), or high (> 3 servings/week). We assessed overall bacterial diversity and presence of specific taxa with respect to HSB intake in each cohort separately and combined in a meta-analysis. Consistently in the two cohorts, we found lower species richness in high HSB consumers (> 3 cans/week) (p = 0.027), and that overall bacterial community profiles differed from those of non-consumers (PERMANOVA p = 0.040). Specifically, presence of a network of commensal bacteria (Lachnospiraceae, Peptostreptococcaceae, and Alloprevotella rava) was less common in high compared to non-consumers, as were other species including Campylobacter showae, Prevotella oulorum, and Mycoplasma faucium. Presence of acidogenic bacteria Bifodobacteriaceae and Lactobacillus rhamnosus was more common in high consumers. Abundance of Fusobacteriales and its genus Leptotrichia, Lachnoanaerobaculum sp., and Campylobacter were lower with higher HSB consumption, and their abundances were correlated. No significant interaction was found for these associations with diabetic status or with microbial markers for caries (S. mutans) and periodontitis (P. gingivalis). Our results suggest that soft drink intake may alter the salivary microbiota, with consistent results across two independent cohorts. The observed perturbations of overrepresented acidogenic bacteria and underrepresented commensal bacteria in high HSB consumers may have implications for oral and systemic disease risk.
PMCID:11167035
PMID: 38862651
ISSN: 2045-2322
CID: 5669042