89 results on '"Hendifar AE"'
Search Results
2. Myositis ossificans: a case report.
- Author
-
Hendifar AE, Johnson D, and Arkfeld DG
- Published
- 2005
3. Hyponatremia in community-acquired pneumonia (CAP): Is it truly a marker for legionella?
- Author
-
Dunbar, LM, Hendifar, AE, Jubelin, BC, Sibley, DET, and McGregor, DW
- Published
- 1999
- Full Text
- View/download PDF
4. Ponsegromab for the Treatment of Cancer Cachexia.
- Author
-
Groarke JD, Crawford J, Collins SM, Lubaczewski S, Roeland EJ, Naito T, Hendifar AE, Fallon M, Takayama K, Asmis T, Dunne RF, Karahanoglu I, Northcott CA, Harrington MA, Rossulek M, Qiu R, and Saxena AR
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Body Weight drug effects, Double-Blind Method, Exercise, Injections, Subcutaneous, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized adverse effects, Appetite drug effects, Cachexia blood, Cachexia drug therapy, Cachexia etiology, Growth Differentiation Factor 15 antagonists & inhibitors, Growth Differentiation Factor 15 blood, Neoplasms blood, Neoplasms complications, Neoplasms drug therapy
- Abstract
Background: Cachexia is a common complication of cancer and is associated with an increased risk of death. The level of growth differentiation factor 15 (GDF-15), a circulating cytokine, is elevated in cancer cachexia. In a small, open-label, phase 1b study involving patients with cancer cachexia, ponsegromab, a humanized monoclonal antibody inhibiting GDF-15, was associated with improved weight, appetite, and physical activity, along with suppressed serum GDF-15 levels., Methods: In this phase 2, randomized, double-blind, 12-week trial, we assigned patients with cancer cachexia and an elevated serum GDF-15 level (≥1500 pg per milliliter) in a 1:1:1:1 ratio to receive ponsegromab at a dose of 100 mg, 200 mg, or 400 mg or to receive placebo, administered subcutaneously every 4 weeks for three doses. The primary end point was the change from baseline in body weight at 12 weeks. Key secondary end points were appetite and cachexia symptoms, digital measures of physical activity, and safety., Results: A total of 187 patients underwent randomization. Of these patients, 40% had non-small-cell lung cancer, 32% had pancreatic cancer, and 29% had colorectal cancer. At 12 weeks, patients in the ponsegromab groups had significantly greater weight gain than those in the placebo group, with a median between-group difference of 1.22 kg (95% credible interval, 0.37 to 2.25) in the 100-mg group, 1.92 (95% credible interval, 0.92 to 2.97) in the 200-mg group, and 2.81 (95% credible interval, 1.55 to 4.08) in the 400-mg group. Improvements were observed across measures of appetite and cachexia symptoms, along with physical activity, in the 400-mg ponsegromab group relative to placebo. Adverse events of any cause were reported in 70% of the patients in the ponsegromab group and in 80% of those in the placebo group., Conclusions: Among patients with cancer cachexia and elevated GDF-15 levels, the inhibition of GDF-15 with ponsegromab resulted in increased weight gain and overall activity level and reduced cachexia symptoms, findings that confirmed the role of GDF-15 as a driver of cachexia. (Funded by Pfizer; ClinicalTrials.gov number, NCT05546476.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
5. Keratin 17 is a prognostic and predictive biomarker in pancreatic ductal adenocarcinoma.
- Author
-
Delgado-Coka LA, Roa-Peña L, Babu S, Horowitz M, Petricoin EF 3rd, Matrisian LM, Blais EM, Marchenko N, Allard FD, Akalin A, Jiang W, Larson BK, Hendifar AE, Picozzi VJ, Choi M, Shroyer KR, and Escobar-Hoyos LF
- Subjects
- Humans, Male, Female, Prognosis, Middle Aged, Aged, Fluorouracil therapeutic use, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Gemcitabine, Immunohistochemistry, Adult, Aged, 80 and over, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal diagnosis, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms mortality, Pancreatic Neoplasms genetics, Pancreatic Neoplasms diagnosis, Biomarkers, Tumor metabolism, Keratin-17 metabolism, Keratin-17 genetics
- Abstract
Objectives: To determine the role of keratin 17 (K17) as a predictive biomarker for response to chemotherapy by defining thresholds of K17 expression based on immunohistochemical tests that could be used to optimize therapeutic intervention for patients with pancreatic ductal adenocarcinoma (PDAC)., Methods: We profiled K17 expression, a hallmark of the basal molecular subtype of PDAC, by immunohistochemistry in 2 cohorts of formalin-fixed, paraffin-embedded PDACs (n = 305). We determined a K17 threshold of expression to optimize prognostic stratification according to the lowest Akaike information criterion and explored the potential relationship between K17 and chemoresistance by multivariate predictive analyses., Results: Patients with advanced-stage, low K17 PDACs treated using 5-fluorouracil (5-FU)-based chemotherapeutic regimens had 3-fold longer survival than corresponding cases treated with gemcitabine-based chemotherapy. By contrast, PDACs with high K17 did not respond to either regimen. The predictive value of K17 was independent of tumor mutation status and other clinicopathologic variables., Conclusions: The detection of K17 in 10% or greater of PDAC cells identified patients with shortest survival. Among patients with low K17 PDACs, 5-FU-based treatment was more likely than gemcitabine-based therapies to extend survival., (© American Society for Clinical Pathology, 2024.)
- Published
- 2024
- Full Text
- View/download PDF
6. Low booster uptake in cancer patients despite health benefits.
- Author
-
Figueiredo JC, Levy J, Choi SY, Xu AM, Merin NM, Hamid O, Lemos T, Nguyen N, Nadri M, Gonzalez A, Mahov S, Darrah JM, Gong J, Paquette RL, Mita AC, Vescio RA, Salvy SJ, Mehmi I, Hendifar AE, Natale R, Tourtellotte WG, Ramanujan VK, Huynh CA, Sobhani K, Reckamp KL, and Merchant AA
- Abstract
Patients with cancer are at increased risk of death from COVID-19 and have reduced immune responses to SARS-CoV2 vaccines, necessitating regular boosters. We performed comprehensive chart reviews, surveys of patients attitudes, serology for SARS-CoV-2 antibodies and T cell receptor (TCR) β sequencing for cellular responses on a cohort of 982 cancer patients receiving active cancer therapy accrued between November-3-2020 and Mar-31-2023. We found that 92 · 3% of patients received the primer vaccine, 70 · 8% received one monovalent booster, but only 30 · 1% received a bivalent booster. Booster uptake was lower under age 50, and among African American or Hispanic patients. Nearly all patients seroconverted after 2+ booster vaccinations (>99%) and improved cellular responses, demonstrating that repeated boosters could overcome poor response to vaccination. Receipt of booster vaccinations was associated with a lower risk of all-cause mortality (HR = 0 · 61, p = 0 · 024). Booster uptake in high-risk cancer patients remains low and strategies to encourage booster uptake are needed., Competing Interests: N.M. holds a consultant or advisory role at Amgen, Kite, Epizyme, TG Therapeutics, ADC Therapeutics, and has research funding from Miltenyi, Teva, and Amgen. J.G. holds a consultant or advisory role at EMD Serono; Elsevier; Exelixis; QED Therapeutics; Natera, Basilea, HalioDx, Eisai, Janssen. O.H. has obtained consulting fees/support meetings/travel/Financial interests in Alkermes, Amgen, Bactonix, Beigene, Bioatla, BMS, Esai, Roche, Genentech, Georgiamune, GigaGen, Grit Bio, GSK, Idera, Immunocore, Incyte, Instilbio, IO Bio, Iovance, Janssen, KSQ, Merck Moderna, Novartis, Obsidian, Pfizer, Regeneron, Sanofi, Seattle Genetics, Tempus, Vial, Zelluna. K.R. holds a consultant or advisory role at Amgen, AstraZeneca, Blueprint, Boehringer Ingelheim, Daiichi Sankyo, EMD Soreno, Genentech, GSK, Janssen, Lilly, Merck KGA, Mirati, Seattle Genetics, Takeda. J.D. holds a consultant or advisory role Kite Pharma and Morphosys. R.V. is on the Speaker’s Bureau for: Amgen, Bristol Myers Squib, Glaxo Smith Klein, Janssen, Karyopharm, and Takeda Pharmaceuticals. A.M. holds a consultant or advisory role at Novartis and Morphosys and has research funding from Amgen and Pfizer., (© 2024 The Authors. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
7. Glutamine Supplementation as an Anticancer Strategy: A Potential Therapeutic Alternative to the Convention.
- Author
-
Muranaka H, Akinsola R, Billet S, Pandol SJ, Hendifar AE, Bhowmick NA, and Gong J
- Abstract
Glutamine, a multifaceted nonessential/conditionally essential amino acid integral to cellular metabolism and immune function, holds pivotal importance in the landscape of cancer therapy. This review delves into the intricate dynamics surrounding both glutamine antagonism strategies and glutamine supplementation within the context of cancer treatment, emphasizing the critical role of glutamine metabolism in cancer progression and therapy. Glutamine antagonism, aiming to disrupt tumor growth by targeting critical metabolic pathways, is challenged by the adaptive nature of cancer cells and the complex metabolic microenvironment, potentially compromising its therapeutic efficacy. In contrast, glutamine supplementation supports immune function, improves gut integrity, alleviates treatment-related toxicities, and improves patient well-being. Moreover, recent studies highlighted its contributions to epigenetic regulation within cancer cells and its potential to bolster anti-cancer immune functions. However, glutamine implementation necessitates careful consideration of potential interactions with ongoing treatment regimens and the delicate equilibrium between supporting normal cellular function and promoting tumorigenesis. By critically assessing the implications of both glutamine antagonism strategies and glutamine supplementation, this review aims to offer comprehensive insights into potential therapeutic strategies targeting glutamine metabolism for effective cancer management.
- Published
- 2024
- Full Text
- View/download PDF
8. The Disproportionate Rise in Pancreatic Cancer in Younger Women Is Due to a Rise in Adenocarcinoma and Not Neuroendocrine Tumors: A Nationwide Time-Trend Analysis Using 2001-2018 United States Cancer Statistics Databases.
- Author
-
Jiang Y, Abboud Y, Liang J, Larson B, Osipov A, Gong J, Hendifar AE, Atkins K, Liu Q, Nissen NN, Li D, Pandol SJ, Lo SK, and Gaddam S
- Abstract
In previous studies, a significant increase in the incidence of pancreatic cancer among younger women compared to men in the United States was noted. However, the specific histopathologic characteristics were not delineated. This population-based study aimed to assess whether this disproportionate rise in pancreatic cancer in younger women was contributed by pancreatic ductal adenocarcinoma (PDAC) or pancreatic neuroendocrine tumors (PanNET). The United States Cancer Statistics (USCS) database was used to identify patients with pancreatic cancer between 2001 and 2018. The results showed that, in younger adults, the incidence of PDAC has increased in women [average annual percentage change (AAPC) = 0.62%], while it has remained stable in men (AAPC = -0.09%). The PDAC incidence rate among women increased at a greater rate compared to men with a statistically significant difference in AAPC ( p < 0.001), with neither identical nor parallel trends. In contrast, cases of PanNET did not demonstrate a statistically significant sex-specific AAPC difference. In conclusion, this study demonstrated that the dramatic increase in the incidence rate of PDAC explains the disproportionate rise in pancreatic cancer incidence in younger women. This prompts further prospective studies to investigate the underlying reasons for these sex-specific disparities in PDAC.
- Published
- 2024
- Full Text
- View/download PDF
9. Cancer-related cells and oncosomes in the liquid biopsy of pancreatic cancer patients undergoing surgery.
- Author
-
Shishido SN, Lin E, Nissen N, Courcoubetis G, Suresh D, Mason J, Osipov A, Hendifar AE, Lewis M, Gaddam S, Pandol S, Kuhn P, and Lo SK
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a five-year survival rate of less than 10% due to its late diagnosis, rapid metastasis, and chemotherapeutic resistance. For a small proportion (10-20%) of early-stage patients however, surgical resection of the pancreatic tumor offers the best chance for survival but the effect of surgery on disease dissemination is unknown. The primary objective of this study was to characterize cellular and acellular blood-based analytes in portal and peripheral blood before pancreatic manipulation, during tumor dissection and immediately after surgical resection to determine the effects of the surgery. This study used the non-enriching third generation High-Definition Single Cell Assay (HDSCA3.0) workflow to investigate heterogeneous circulating rare cell population in the blood. Blood from both sites taken before surgical manipulation of the pancreas had significantly greater incidence of total rare cellular and acellular analytes than normal donor samples. Post-surgery portal and peripheral blood had significantly greater incidence of specific cellular and acellular subtypes compared to the matched pre- and during-surgery samples. Our results reveal that in patients with PDAC liquid biopsy analytes are increased in both the portal and peripheral blood; portal blood contains a higher frequency of analytes than in the peripheral blood; total analytes in the portal and peripheral blood samples were significantly associated with the tumor volume and pathological T stage; and the surgical procedure increased the blood levels of circulating cellular and acellular analytes, but not Epi.CTCs or Mes.CTCs. This study demonstrates liquid biopsy's utility in monitoring patients with PDAC with surgically resectable disease., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Patterns of Failure in Patients With Borderline Resectable/Locally Advanced Pancreatic Cancer After Preoperative Chemotherapy and Stereotactic Body Radiation Therapy.
- Author
-
Chung EM, Lu DJ, Nguyen AT, Hendifar AE, Nissen NN, Gong J, Osipov A, Gangi A, Attiyeh MA, Atkins KM, and Kamrava M
- Abstract
Purpose: The role of preoperative stereotactic body radiation therapy (SBRT) in pancreatic cancer is controversial, and questions regarding the optimal dose and radiation treatment field remain. To better inform future investigations of SBRT dose and radiation fields, we evaluated the patterns of failure in patients with borderline resectable/locally advanced pancreatic cancer (BR/LAPC) after preoperative chemotherapy and SBRT in patients who underwent surgical resection., Methods and Materials: We performed a single-institution retrospective review of consecutive patients treated from September 2017 to January 2022 with BR/LAPC. Patients who underwent preoperative chemotherapy and SBRT followed by surgical resection were reviewed. SBRT was delivered to a dose of 33 Gy in 5 fractions. Kaplan-Meier overall survival and progression-free survival estimates were calculated., Results: In total, 18 patients (12 BRPC, 6 LAPC) were included. Median age was 69 years (range 41-84 years). Median follow-up was 30 months (range 13-59 months). Seventeen patients (94%) had a R0 resection and 13 (72%) underwent vascular reconstruction. Median overall survival and progression-free survival was 42 months (range 13-59 months) and 23 months (range 1-45 months), respectively. In total, 61% (11/18) patients experienced progression at any point during follow-up. Of the patients who experienced recurrence, 27% (3/11) experienced local progression as component of their first recurrence, whereas 100% (11/11) experienced distant progression as a component of their first recurrence. When examining all recurrences that occurred at any point in follow-up, 28% (5/18) of patients experienced local or locoregional recurrence and 61% (11/18) experienced distant progression., Conclusions: Local control and margin negative resection rates were excellent with preoperative chemotherapy and nondose-escalated SBRT in surgically resected patients with BR/LAPC. Distant recurrence was the predominant site of failure with lower incidences of isolated locoregional recurrences. Additional research is needed to determine the ideal treatment volume and patients who may benefit from dose escalation., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
11. Recent advances in the management of hepatocellular carcinoma.
- Author
-
Sankar K, Gong J, Osipov A, Miles SA, Kosari K, Nissen NN, Hendifar AE, Koltsova EK, and Yang JD
- Subjects
- Humans, Liver Cirrhosis complications, Carcinoma, Hepatocellular therapy, Carcinoma, Hepatocellular etiology, Liver Neoplasms complications, Hepatitis B complications
- Abstract
Liver cancer remains a challenge of global health, being the 4th leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, and is usually precipitated by chronic viral infections (hepatitis B and C), non-alcoholic steatohepatitis, heavy alcohol use, and other factors which may lead to chronic inflammation and cirrhosis of the liver. There have been significant advances in the systemic treatment options for HCC over the past decades, with several approvals of both immune checkpoint inhibitors and tyrosine kinase inhibitors in patients with preserved liver function. These advances have led to improvement in survival outcomes, with expected survival of greater than 18 months, in those with sensitive tumors, adequate liver function, and those functionally fit to receive sequential therapies. Several ongoing and promising trials are now evaluating combinational strategies with novel systemic agents and combinations of systemic therapy with locoregional therapy. In view of these trials, further advances in the treatment of HCC are foreseen in the near future.
- Published
- 2024
- Full Text
- View/download PDF
12. Socioeconomic disparities in patients with small bowel neuroendocrine tumors.
- Author
-
DiPeri TP, Manguso N, Gong J, Atkins KM, Hendifar AE, and Gangi A
- Subjects
- Humans, Retrospective Studies, Socioeconomic Disparities in Health, Socioeconomic Factors, Healthcare Disparities, Neuroendocrine Tumors therapy
- Abstract
Background and Objectives: Demographic and socioeconomic disparities affect cancer specific outcomes in numerous malignancies, but the impact of these for patients with small bowel neuroendocrine tumors (SBNETs) is not well understood. The primary objective was to investigate the impact of demographic and socioeconomic factors on overall survival (OS) for patients with SBNETs., Methods: We performed a retrospective cohort study utilizing the National Cancer Database to assess patients diagnosed with SBNET between 2004 and 2015. Patients were stratified by demographics, socioeconomic factors, insurance status, and place of living., Results: The 5-year OS for the entire cohort was 78.5%. The 5-year survival was worse in patients with lower income (p < 0.0001), lower education (p < 0.0001), not in proximity to a metro area (p = 0.0004), and treatment at a community cancer center (p < 0.0001). Adjusting for age and sex, factors associated with worse OS were lower income (<$38 000) (hazard ratio [HR]: 1.16, 95% confidence interval [CI]: 1.04-1.28), lower education (>20% no HSD) (HR: 1.14, 95% CI: 1.02-1.26), no insurance (HR: 1.66, 95% CI: 1.33-2.06), and not living in proximity to a metro area (HR: 1.27, 95% CI: 1.10-1.47)., Conclusions: Patient demographics and socioeconomic factors play an important role in survival of patients with SBNETs, specifically proximity to a metro area, median income, education level, and type of treatment center. Strategies to improve access to care must be considered in this at-risk population., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
13. Low booster uptake in cancer patients despite health benefits.
- Author
-
Figueiredo JC, Levy J, Choi SY, Xu AM, Merin NM, Hamid O, Lemos T, Nguyen N, Nadri M, Gonzalez A, Mahov S, Darrah JM, Gong J, Paquette RL, Mita AC, Vescio RA, Salvy SJ, Mehmi I, Hendifar AE, Natale R, Tourtellotte WG, Krishnan Ramanujan V, Huynh CA, Sobhani K, Reckamp KL, and Merchant AA
- Abstract
Patients with cancer are at increased risk of death from COVID-19 and have reduced immune responses to SARS-CoV2 vaccines, necessitating regular boosters. We performed comprehensive chart reviews, surveys of patients attitudes, serology for SARS-CoV-2 antibodies and T-cell receptor (TCR) β sequencing for cellular responses on a cohort of 982 cancer patients receiving active cancer therapy accrued between November-3-2020 and Mar-31-2023. We found that 92·3% of patients received the primer vaccine, 70·8% received one monovalent booster, but only 30·1% received a bivalent booster. Booster uptake was lower under age 50, and among African American or Hispanic patients. Nearly all patients seroconverted after 2+ booster vaccinations (>99%) and improved cellular responses, demonstrating that repeated boosters could overcome poor response to vaccination. Receipt of booster vaccinations was associated with a lower risk of all-cause mortality (HR=0·61, P=0·024). Booster uptake in high-risk cancer patients remains low and strategies to encourage booster uptake are needed., Highlights: COVID-19 booster vaccinations increase antibody levels and maintain T-cell responses against SARS-CoV-2 in patients receiving various anti-cancer therapiesBooster vaccinations reduced all-cause mortality in patientsA significant proportion of patients remain unboosted and strategies are needed to encourage patients to be up-to-date with vaccinations.
- Published
- 2023
- Full Text
- View/download PDF
14. Pancreatoblastoma in Elderly Adults: Report of Two Patients.
- Author
-
Kulak O, Osipov A, Hendifar AE, Nissen NN, Cox BK, Hruban RH, and Hutchings DA
- Subjects
- Humans, Adult, Child, Aged, Aged, 80 and over, Pancreas pathology, Pancreatectomy, Cell Differentiation, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Introduction . Pancreatoblastoma is a rare malignant epithelial neoplasm of the pancreas, which often shows multiple lines of differentiation, but is defined by neoplastic cells with acinar differentiation and characteristic squamoid nests. Pediatric patients are most commonly affected, and although a subset is known to occur in adults, the diagnosis is rarely considered in elderly adults. Methods. The clinicopathologic features of two cases of pancreatoblastoma in elderly patients were examined. Results. Two patients (age 80 and 81 years) presented with pancreatoblastoma, including one with early-stage pancreatic disease and one with liver metastasis. Biopsies and one pancreatic resection specimen showed characteristic histomorphologic features, including prominent acinar differentiation and abundant squamoid nests. Both cases had complete loss of SMAD4 (DPC4) immunolabeling. Next generation sequencing was performed on one case and revealed copy number loss of chromosome 11p and 9p21 ( CDKN2A/B ) and pathogenic or likely pathogenic variants in APC , SMAD4 , and PIK3CA . The APC and SMAD4 variants occurred at allele frequencies suggestive of germline mutations, raising the possibility that this patient may have an inherited cancer predisposition syndrome. Conclusions. We present two cases which extend the upper age limit for reported pancreatoblastoma, including one with genetic findings suggestive of an inherited cancer predisposition syndrome.
- Published
- 2023
- Full Text
- View/download PDF
15. Combination L-Glutamine with Gemcitabine and Nab-Paclitaxel in Treatment-Naïve Advanced Pancreatic Cancer: The Phase I GlutaPanc Study Protocol.
- Author
-
Gong J, Osipov A, Lorber J, Tighiouart M, Kwan AK, Muranaka H, Akinsola R, Billet S, Levi A, Abbas A, Davelaar J, Bhowmick N, and Hendifar AE
- Abstract
Advanced pancreatic cancer is underscored by progressive therapeutic resistance and a dismal 5-year survival rate of 3%. Preclinical data demonstrated glutamine supplementation, not deprivation, elicited antitumor effects against pancreatic ductal adenocarcinoma (PDAC) alone and in combination with gemcitabine in a dose-dependent manner. The GlutaPanc phase I trial is a single-arm, open-label clinical trial investigating the safety of combination L-glutamine, gemcitabine, and nab-paclitaxel in subjects ( n = 16) with untreated, locally advanced unresectable or metastatic pancreatic cancer. Following a 7-day lead-in phase with L-glutamine, the dose-finding phase via Bayesian design begins with treatment cycles lasting 28 days until disease progression, intolerance, or withdrawal. The primary objective is to establish the recommended phase II dose (RP2D) of combination L-glutamine, gemcitabine, and nab-paclitaxel. Secondary objectives include safety of the combination across all dose levels and preliminary evidence of antitumor activity. Exploratory objectives include evaluating changes in plasma metabolites across multiple time points and changes in the stool microbiome pre and post L-glutamine supplementation. If this phase I clinical trial demonstrates the feasibility of L-glutamine in combination with nab-paclitaxel and gemcitabine, we would advance the development of this combination as a first-line systemic option in subjects with metastatic pancreatic cancer, a high-risk subgroup desperately in need of additional therapies.
- Published
- 2023
- Full Text
- View/download PDF
16. TOPAZ-1: a new standard of care for advanced biliary tract cancers?
- Author
-
Ebia MI, Sankar K, Osipov A, Hendifar AE, and Gong J
- Subjects
- Humans, Standard of Care, Antineoplastic Combined Chemotherapy Protocols, Bile Duct Neoplasms, Biliary Tract Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
17. Immunotherapy for deficient mismatch repair (dMMR) pancreatic ductal adenocarcinoma.
- Author
-
Ebia MI, Hitchins MP, and Hendifar AE
- Abstract
Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jgo.amegroups.com/article/view/10.21037/jgo-23-12/coif). The authors have no conflicts of interest to declare.
- Published
- 2023
- Full Text
- View/download PDF
18. Randomized Study of Temozolomide or Temozolomide and Capecitabine in Patients With Advanced Pancreatic Neuroendocrine Tumors (ECOG-ACRIN E2211).
- Author
-
Kunz PL, Graham NT, Catalano PJ, Nimeiri HS, Fisher GA, Longacre TA, Suarez CJ, Martin BA, Yao JC, Kulke MH, Hendifar AE, Shanks JC, Shah MH, Zalupski MM, Schmulbach EL, Reidy-Lagunes DL, Strosberg JR, O'Dwyer PJ, and Benson AB 3rd
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Capecitabine therapeutic use, Dacarbazine therapeutic use, Prospective Studies, Retrospective Studies, Temozolomide therapeutic use, Treatment Outcome, Neuroendocrine Tumors drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: Patients with advanced pancreatic neuroendocrine tumors (NETs) have few treatment options that yield objective responses. Retrospective and small prospective studies suggest that capecitabine and temozolomide are associated with high response rates (RRs) and long progression-free survival (PFS)., Patients and Methods: E2211 was a multicenter, randomized, phase II trial comparing temozolomide versus capecitabine/temozolomide in patients with advanced low-grade or intermediate-grade pancreatic NETs. Key eligibility criteria included progression within the preceding 12 months and no prior temozolomide, dimethyl-triazeno-imidazole-carboxamide or dacarbazine, capecitabine or fluorouracil. The primary end point was PFS; secondary endpoints were overall survival, RR, safety, and methylguanine methyltransferase (MGMT) by immunohistochemistry and promoter methylation., Results: A total of 144 patients were enrolled between April 2013 and March 2016 to temozolomide (n = 72) or capecitabine and temozolomide (n = 72); the primary analysis population included 133 eligible patients. At the scheduled interim analysis in January 2018, the median PFS was 14.4 months for temozolomide versus 22.7 months for capecitabine/temozolomide (hazard ratio = 0.58), which was sufficient to reject the null hypothesis for the primary end point (stratified log-rank P = .022). In the final analysis (May 2021), the median overall survival was 53.8 months for temozolomide and 58.7 months for capecitabine/temozolomide (hazard ratio = 0.82, P = .42). MGMT deficiency was associated with response., Conclusion: The combination of capecitabine/temozolomide was associated with a significant improvement in PFS compared with temozolomide alone in patients with advanced pancreatic NETs. The median PFS and RR observed with capecitabine/temozolomide are the highest reported in a randomized study for pancreatic NETs. MGMT deficiency was associated with response, and although routine MGMT testing is not recommended, it can be considered for select patients in need of objective response (ClinicalTrials.gov identifier: NCT01824875).
- Published
- 2023
- Full Text
- View/download PDF
19. Multitasking dynamic contrast enhanced magnetic resonance imaging can accurately differentiate chronic pancreatitis from pancreatic ductal adenocarcinoma.
- Author
-
Wang N, Gaddam S, Xie Y, Christodoulou AG, Wu C, Ma S, Fan Z, Wang L, Lo S, Hendifar AE, Pandol SJ, and Li D
- Abstract
Background and Aims: Accurate differentiation of chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC) is an area of unmet clinical need. In this study, a novel Multitasking dynamic contrast enhanced (DCE) magnetic resonance imaging (MRI) technique was used to quantitatively evaluate the microcirculation properties of pancreas in CP and PDAC and differentiate between them., Methods: The Multitasking DCE technique was able to acquire one 3D image per second during the passage of MRI contrast agent, allowing the quantitative estimation of microcirculation properties of tissue, including blood flow F
p , plasma volume fraction vp , transfer constant Ktrans , and extravascular extracellular volume fraction ve . Receiver operating characteristic (ROC) analysis was performed to differentiate the CP pancreas, PDAC pancreas, normal control pancreas, PDAC tumor, PDAC upstream, and PDAC downstream. ROCs from quantitative analysis and conventional analysis were compared., Results: Fourteen PDAC patients, 8 CP patients and 20 healthy subjects were prospectively recruited. The combination of Fp , vp , Ktrans , and ve can differentiate CP versus PDAC pancreas with good AUC (AUC [95% CI] = 0.821 [0.654 - 0.988]), CP versus normal pancreas with excellent AUC (1.000 [1.000 - 1.000]), PDAC pancreas versus normal pancreas with excellent AUC (1.000 [1.000 - 1.000]), CP versus PDAC tumor with excellent AUC (1.000 [1.000 - 1.000]), CP versus PDAC downstream with excellent AUC (0.917 [0.795 - 1.000]), and CP versus PDAC upstream with fair AUC (0.722 [0.465 - 0.980]). This quantitative analysis outperformed conventional analysis in differentiation of each pair., Conclusion: Multitasking DCE MRI is a promising clinical tool that is capable of unbiased quantitative differentiation between CP from PDAC., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Wang, Gaddam, Xie, Christodoulou, Wu, Ma, Fan, Wang, Lo, Hendifar, Pandol and Li.)- Published
- 2023
- Full Text
- View/download PDF
20. A phase II study of sapanisertib (TAK-228) a mTORC1/2 inhibitor in rapalog-resistant advanced pancreatic neuroendocrine tumors (PNET): ECOG-ACRIN EA2161.
- Author
-
Rajdev L, Lee JW, Libutti SK, Benson AB, Fisher GA Jr, Kunz PL, Hendifar AE, Catalano P, and O'Dwyer PJ
- Subjects
- Humans, Mechanistic Target of Rapamycin Complex 1 metabolism, MTOR Inhibitors, Protein Kinase Inhibitors adverse effects, Sirolimus, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology, Neuroectodermal Tumors, Primitive drug therapy
- Abstract
This was a two-stage phase II trial of a mTORC1/2 inhibitor (mTORC: mammalian target of rapamycin complex) Sapanisertib (TAK228) in patients with rapalog-resistant pancreatic neuroendocrine tumors (PNETs) (NCT02893930). Approved rapalogs such as everolimus inhibit mTORC1 and have limited clinical activity, possibly due to compensatory feedback loops. Sapanisertib addresses the potential for incomplete inhibition of the mTOR pathway through targeting of both mTORC1 and mTORC2, and thus to reverse resistance to earlier rapamycin analogues. In stage 1, patients received sapanisertib 3 mg by mouth once daily on a continuous dosing schedule in 28-day cycle. This trial adopted a two-stage design with the primary objective of evaluating objective tumor response. The first stage would recruit 13 patients in order to accrue 12 eligible and treated patients. If among the 12 eligible patients at least 1 patient had an objective response to therapy, the study would move to the second stage of accrual where 25 eligible and treated patients would be enrolled. This study activated on February 1, 2017, the required pre-determined number of patients (n = 13) had entered by November 5, 2018 for the first stage response evaluation. The accrual of this trial was formally terminated on December 27, 2019 as no response had been observed after the first stage accrual. Treatment-related grade 3 adverse events were reported in eight (61%) patients with hyperglycemia being the most frequent, in three patients (23%). Other toxicities noted in the trial included fatigue, rash diarrhea, nausea, and vomiting. The median PFS was 5.19 months (95% CI [3.84, 9.30]) and the median OS was 20.44 months (95% CI [5.65, 22.54]). Due to the lack of responses in Stage 1 of the study, the study did not proceed to stage 2. Thus the potential to reverse resistance was not evident., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
21. Tumor hyaluronan as a novel biomarker in non-small cell lung cancer: A retrospective study.
- Author
-
Gong J, Guan M, Kim H, Moshayedi N, Mehta S, Cook-Wiens G, Larson BK, Zhou J, Patel R, Lapite I, Placencio-Hickok VR, Tuli R, Natale RB, and Hendifar AE
- Subjects
- Humans, Hyaluronic Acid metabolism, Retrospective Studies, Neoplasm Recurrence, Local, Biomarkers, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms pathology, Adenocarcinoma metabolism, Carcinoma, Squamous Cell
- Abstract
Introduction: Hyaluronan (HA) accumulation is associated with tumorigenesis and aggressive tumor behavior., Aims: We investigated the biomarker potential of HA in non-small cell lung cancer (NSCLC)., Methods: HA levels were scored using affinity histochemistry in 137 NSCLC samples stratified by HA score ≤10, 11-20, 21-30, and >30 with HA-high defined as ≥25% expression in the extracellular matrix (ECM) of the tumor surface area. Overall survival (OS) and time to progression from initiation of taxane therapy (TTP) were compared using log-rank tests based on HA score., Results: Of 122 patients with recurrent/metastatic NSCLC, 93 had mean HA scores that were not significantly different across clinicopathologic variables. Frequency of HA-high tumors did not differ by histology (34/68 adenocarcinomas vs. 12/25 squamous tumors, Fisher's p = 1.0000). Median OS for recurrent/metastatic adenocarcinoma was 35.5 months (95%, 23.6-50.3) vs. 17.9 months for squamous (95%, 12.7-37.0, log-rank test, p = 0.0165). OS was not significantly different by HA quartiles, high or low (<25) HA score and tumor histology, and HA biopsy site (all p > 0.05). Median TTP ( n = 98) significantly differed by HA quartile (2.8 months for HA score ≤10; 5.0 months for 11-20; 7.9 months for 21-30; 3.9 months for >30, p = 0.0265). Improved TTP trended in HA-high over HA-low tumors ( n = 98, p = 0.0911)., Conclusion: In this NSCLC cohort, tumor HA level represents a potential biomarker for TTP, which remains a cornerstone of NSCLC therapy. Further validation is warranted to identify the HA accumulation threshold associated with clinical benefit.
- Published
- 2022
- Full Text
- View/download PDF
22. Dual Stromal Targeting Sensitizes Pancreatic Adenocarcinoma for Anti-Programmed Cell Death Protein 1 Therapy.
- Author
-
Blair AB, Wang J, Davelaar J, Baker A, Li K, Niu N, Wang J, Shao Y, Funes V, Li P, Pachter JA, Maneval DC, Dezem F, Plummer J, Chan KS, Gong J, Hendifar AE, Pandol SJ, Burkhart R, Zhang Y, Zheng L, and Osipov A
- Subjects
- Humans, Mice, Animals, Hyaluronoglucosaminidase pharmacology, Hyaluronoglucosaminidase therapeutic use, Hyaluronic Acid, Focal Adhesion Protein-Tyrosine Kinases, Cytokines pharmacology, Cell Death, Polyethylene Glycols therapeutic use, Tumor Microenvironment, Pancreatic Neoplasms, Pancreatic Neoplasms pathology, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal genetics, Liver Neoplasms drug therapy
- Abstract
Background & Aims: The stroma in pancreatic ductal adenocarcinoma (PDAC) contributes to its immunosuppressive nature and therapeutic resistance. Herein we sought to modify signaling and enhance immunotherapy efficacy by targeting multiple stromal components through both intracellular and extracellular mechanisms., Methods: A murine liver metastasis syngeneic model of PDAC was treated with focal adhesion kinase inhibitor (FAKi), anti-programmed cell death protein 1 (PD-1) antibody, and stromal hyaluronan (HA) degradation by PEGylated recombinant human hyaluronidase (PEGPH20) to assess immune and stromal modulating effects of these agents and their combinations., Results: The results showed that HA degradation by PEGPH20 and reduction in phosphorylated FAK expression by FAKi leads to improved survival in PDAC-bearing mice treated with anti-PD-1 antibody. HA degradation in combination with FAKi and anti-PD-1 antibody increases T-cell infiltration and alters T-cell phenotype toward effector memory T cells. FAKi alters the expression of T-cell modulating cytokines and leads to changes in T-cell metabolism and increases in effector T-cell signatures. HA degradation in combination with anti-PD-1 antibody and FAKi treatments reduces granulocytes, including granulocytic- myeloid-derived suppressor cells and decreases C-X-C chemokine receptor type 4 (CXCR4)-expressing myeloid cells, particularly the CXCR4-expressing granulocytes. Anti-CXCR4 antibody combined with FAKi and anti-PD-1 antibody significantly decreases metastatic rates in the PDAC liver metastasis model., Conclusions: This represents the first preclinical study to identify synergistic effects of targeting both intracellular and extracellular components within the PDAC stroma and supports testing anti-CXCR4 antibody in combination with FAKi as a PDAC treatment strategy., (Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
23. Exceptional Response to Erdafitinib in FGFR2-Mutated Metastatic Pancreatic Ductal Adenocarcinoma.
- Author
-
Ng CF, Glaspy J, Placencio-Hickok VR, Thomassian S, Gong J, Osipov A, Hendifar AE, and Moshayedi N
- Subjects
- Humans, Pyrazoles, Quinoxalines, Receptor, Fibroblast Growth Factor, Type 2 genetics, Receptor, Fibroblast Growth Factor, Type 2 therapeutic use, Tumor Microenvironment, Pancreatic Neoplasms, Adenocarcinoma genetics, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Despite advances in cancer therapeutics, pancreatic ductal adenocarcinoma (PDAC) remains among the deadliest malignancies, with a poor prognosis at time of diagnosis. Research in PDAC has suggested that adaptive signaling in the tumor microenvironment may promote tumor proliferation and survival. Several FGFR fusion genes-specifically FGFR2-are involved with the creation and progression of cancer. These mutations are found in a variety of cancer types. This report presents a unique case of a young patient with stage IV PDAC with a known FGFR2 fusion. This molecular alteration afforded a remarkable response to FGFR inhibitor therapy, erdafitinib, after the patient experienced disease progression on multiple chemotherapy regimens.
- Published
- 2022
- Full Text
- View/download PDF
24. Molecular Characterization of KRAS Wild-type Tumors in Patients with Pancreatic Adenocarcinoma.
- Author
-
Philip PA, Azar I, Xiu J, Hall MJ, Hendifar AE, Lou E, Hwang JJ, Gong J, Feldman R, Ellis M, Stafford P, Spetzler D, Khushman MM, Sohal D, Lockhart AC, Weinberg BA, El-Deiry WS, Marshall J, Shields AF, and Korn WM
- Subjects
- Aged, DNA Helicases genetics, Female, Humans, Male, Microsatellite Instability, Mutation, Nuclear Proteins genetics, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Transcription Factors genetics, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Purpose: KRAS mutation (MT) is a major oncogenic driver in pancreatic ductal adenocarcinoma (PDAC). A small subset of PDACs harbor KRAS wild-type (WT). We aim to characterize the molecular profiles of KRAS WT PDAC to uncover new pathogenic drivers and offer targeted treatments., Experimental Design: Tumor tissue obtained from surgical or biopsy material was subjected to next-generation DNA/RNA sequencing, microsatellite instability (MSI) and mismatch repair status determination., Results: Of the 2,483 patients (male 53.7%, median age 66 years) studied, 266 tumors (10.7%) were KRAS WT. The most frequently mutated gene in KRAS WT PDAC was TP53 (44.5%), followed by BRAF (13.0%). Multiple mutations within the DNA-damage repair (BRCA2, ATM, BAP1, RAD50, FANCE, PALB2), chromatin remodeling (ARID1A, PBRM1, ARID2, KMT2D, KMT2C, SMARCA4, SETD2), and cell-cycle control pathways (CDKN2A, CCND1, CCNE1) were detected frequently. There was no statistically significant difference in PD-L1 expression between KRAS WT (15.8%) and MT (17%) tumors. However, KRAS WT PDAC were more likely to be MSI-high (4.7% vs. 0.7%; P < 0.05), tumor mutational burden-high (4.5% vs. 1%; P < 0.05), and exhibit increased infiltration of CD8+ T cells, natural killer cells, and myeloid dendritic cells. KRAS WT PDACs exhibited gene fusions of BRAF (6.6%), FGFR2 (5.2%), ALK (2.6%), RET (1.3%), and NRG1 (1.3%), as well as amplification of FGF3 (3%), ERBB2 (2.2%), FGFR3 (1.8%), NTRK (1.8%), and MET (1.3%). Real-world evidence reveals a survival advantage of KRAS WT patients in overall cohorts as well as in patients treated with gemcitabine/nab-paclitaxel or 5-FU/oxaliplatin., Conclusions: KRAS WT PDAC represents 10.7% of PDAC and is enriched with targetable alterations, including immuno-oncologic markers. Identification of KRAS WT patients in clinical practice may expand therapeutic options in a clinically meaningful manner., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
25. Claudin-18.
- Author
-
Wong MT, Singhi AD, Larson BK, Huynh CAT, Balzer BL, Burch M, Dhall D, Gangi A, Gong J, Guindi M, Hendifar AE, Kim SA, de Peralta-Venturina M, and Waters KM
- Subjects
- Humans, Esophagogastric Junction pathology, Metaplasia pathology, Hyperplasia pathology, Claudins, Stomach Neoplasms pathology, Gastritis pathology, Adenocarcinoma pathology, Precancerous Conditions pathology, Neuroendocrine Tumors pathology
- Abstract
Context.—: Claudin-18 is expressed in some gastric cancers. Clinical trials are evaluating it as a therapeutic target., Objectives.—: To evaluate claudin-18 expression in intestinal metaplasia, dysplasia, and adenocarcinoma of the distal esophagus/gastroesophageal junction and stomach and to evaluate claudin-18 expression in gastric and nongastric neuroendocrine tumors as a marker of gastric origin., Design.—: Samples included gastroesophageal junction with intestinal metaplasia (n = 40), dysplasia (n = 54), and adenocarcinoma (n = 20) and stomach with intestinal metaplasia (n = 79), dysplasia (n = 43), and adenocarcinoma (n = 25). Additionally, gastric (n = 40) and nongastric (n = 322) neuroendocrine tumors were included. Claudin-18 expression was evaluated for any staining as positive and by meeting clinical trial inclusion criteria (≥2+ intensity in ≥50% of tumor)., Results.—: Claudin-18 staining was not significantly different across dysplasia categories in the gastroesophageal junction (P = .11) or stomach (P = .12). The rate of positive staining was higher in gastroesophageal junction than stomach for intestinal metaplasia (37 of 40 [92.5%] versus 37 of 79 [46.8%]; P < .001) and high-grade dysplasia (33 of 38 [86.8%] versus 9 of 16 [56.3%]; P = .03). Intestinal metaplasia showed staining in 7 of 37 autoimmune gastritis samples (18.9%) compared with 30 of 42 samples without autoimmune gastritis (71.4%) (P < .001). Adenocarcinoma showed similar staining in gastroesophageal junction (15 of 20; 75.0%) and stomach (17 of 25; 68.0%) (P = .85). Eighty percent (32 of 40) of gastric neuroendocrine tumors were positive for claudin-18 expression, with 57.5% (23 of 40) meeting clinical trial inclusion criteria. Comparatively, 0.62% (2 of 322) of nongastric neuroendocrine tumors showed staining (P < .001)., Conclusions.—: Claudin-18 staining was similar in intestinal metaplasia, dysplasia, and adenocarcinoma. Claudin-18 was negative in most cases of intestinal metaplasia in autoimmune gastritis, indicating that intestinal metaplasia in this setting may differ from other forms. Claudin-18 was sensitive and specific for gastric origin in neuroendocrine tumors.
- Published
- 2022
- Full Text
- View/download PDF
26. Chemical Exchange Saturation Transfer for Pancreatic Ductal Adenocarcinoma Evaluation.
- Author
-
Wang L, Zhou Z, Gaddam S, Wang N, Xie Y, Deng Z, Fan Z, Christodoulou AG, Han F, Lo SK, Wachsman AM, Hendifar AE, Jiang T, Pandol SJ, and Li D
- Subjects
- Humans, Magnetic Resonance Imaging methods, Pancreatic Ducts pathology, Sensitivity and Specificity, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal diagnostic imaging, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms pathology
- Abstract
Objectives: The aims of the study are to evaluate the feasibility of using pH-sensitive magnetic resonance imaging, chemical exchange saturation transfer (CEST) in pancreatic imaging and to differentiate pancreatic ductal adenocarcinoma (PDAC) with the nontumor pancreas (upstream and downstream) and normal control pancreas., Methods: Sixteen CEST images with PDAC and 12 CEST images with normal volunteers were acquired and magnetization transfer ratio with asymmetric analysis were measured in areas of PDAC, upstream, downstream, and normal control pancreas. One-way analysis of variance and receiver operating characteristic curve were used to differentiate tumor from nontumor pancreas., Results: Areas with PDAC showed higher signal intensity than upstream and downstream on CEST images. The mean (standard deviation) values of magnetization transfer ratio with asymmetric analysis were 0.015 (0.034), -0.044 (0.030), -0.019 (0.027), and -0.037 (0.031), respectively, in PDAC area, upstream, downstream, and nontumor area in patient group and -0.008 (0.024) in normal pancreas. Significant differences were found between PDAC and upstream ( P < 0.001), between upstream and normal pancreas ( P = 0.04). Area under curve is 0.857 in differentiating PDAC with nontumor pancreas., Conclusions: pH-sensitive CEST MRI is feasible in pancreatic imaging and can be used to differentiate PDAC from nontumor pancreas. This provides a novel metabolic imaging method in PDAC., Competing Interests: The authors declare no conflict of interest., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. Addressing unmet needs for people with cancer cachexia: recommendations from a multistakeholder workshop.
- Author
-
Garcia JM, Dunne RF, Santiago K, Martin L, Birnbaum MJ, Crawford J, Hendifar AE, Kochanczyk M, Moravek C, Piccinin D, Picozzi V, Roeland EJ, Selig WKD, and Zimmers TA
- Subjects
- Humans, Surveys and Questionnaires, Cachexia etiology, Cachexia therapy, Neoplasms complications, Neoplasms therapy
- Published
- 2022
- Full Text
- View/download PDF
28. Best Practices for the Coordinated Care of Patients With Neuroendocrine Tumors Undergoing Peptide Receptor Radionuclide Therapy.
- Author
-
Hendifar AE, Mehr SH, and McHaffie DR
- Subjects
- Adult, Humans, Intestinal Neoplasms, Octreotide, Pancreatic Neoplasms, Radioisotopes therapeutic use, Receptors, Somatostatin, Somatostatin, Stomach Neoplasms, Neuroendocrine Tumors radiotherapy, Organometallic Compounds therapeutic use
- Abstract
Abstract: Neuroendocrine tumors (NETs) are rare, diverse malignancies; approximately two thirds originate in the gastrointestinal tract and pancreas and are known as gastroenteropancreatic NET. Most cases are diagnosed in the advanced or metastatic setting and overexpress somatostatin receptors. Recommended first-line treatment is somatostatin analogs; however, disease progression is common. [177Lu]Lu-DOTA-TATE is a radiolabeled peptide receptor radionuclide therapy (PRRT) indicated for the treatment of adult patients with somatostatin receptor-positive foregut, midgut, and hindgut gastroenteropancreatic NETs and progression on first-line somatostatin analogs. Many primary oncology practices may lack the staff, expertise, and infrastructure to treat patients with PRRT and primary oncologists may therefore refer their patients to a NET specialist at a tertiary center for treatment. Given the amount of organization required, PRRT treatment may seem to be complex; however, this process will be managed by a care coordinator who acts as a consistent point of contact for primary physicians regarding the care of their patients and ensures blood tests and scans are scheduled. In this article, we share our opinions, procedures, workflow, best practice, and roles and responsibilities when caring for patients receiving [177Lu]Lu-DOTA-TATE and focus on the role of the primary oncologist before, during, and after PRRT treatment., Competing Interests: A.E.H. has consultancy or advisory role in Ipsen, Novartis. The other authors declare no conflict of interest., (Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
29. Hyaluronan heterogeneity in pancreatic ductal adenocarcinoma: Primary tumors compared to sites of metastasis.
- Author
-
Placencio-Hickok VR, Lauzon M, Moshayedi N, Guan M, Kim S, Nissen N, Lo S, Pandol S, Larson BK, Gong J, Hendifar AE, and Osipov A
- Subjects
- Adjuvants, Immunologic, Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal pathology, Humans, Liver Neoplasms, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Prognosis, Pancreatic Neoplasms, Biomarkers, Tumor metabolism, Carcinoma, Pancreatic Ductal diagnosis, Hyaluronic Acid metabolism, Pancreatic Neoplasms diagnosis
- Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with poor survival. The dense desmoplastic stroma in PDAC contributes to treatment resistance. Among the components comprising the tumor stroma, hyaluronan (HA) has been demonstrated to play a critical role in tumor progression and survival. Previous preliminary studies have suggested differences in HA expression in primary and metastatic foci of PDAC. However, the effects of treatment and location of HA expression as a biomarker signature remain unknown; this study sought to compare HA expression in primary and metastatic sites of PDAC., Methods: Tissue from primary and metastatic PDACs were obtained from Cedars-Sinai Medical Center along with associated clinical data. Tissue slides were stained for H&E, HA, and CD44. Associations between HA levels and the evaluated variables were examined including progression free survival and overall survival., Results: HA score was significantly higher in primary PDACs compared to sites of metastases (p = 0.0148). Within the metastases, HA score was significantly higher in liver metastases compared to metastases at other sites (p = 0.0478). In the treatment-naive liver metastasis cohort, patients with HA high status had decreased progression free survival and overall survival compared to patients with HA low status (p = 0.0032 and p = 0.0478, respectively)., Conclusions: HA score is variable between primary PDAC, PDAC metastatic to the liver, and PDAC metastatic to other sites. Within liver metastases, patients with HA high status had decreased progression free survival and overall survival compared to patients with HA low status. HA levels can serve as a potential biomarker to guide pancreatic cancer treatments and trial design for agents targeting the stroma., (Copyright © 2021 IAP and EPC. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Correction to: Midgut Neuroendocrine Tumors with Liver-only Metastases: Benefit of Primary Tumor Resection.
- Author
-
Gangi A, Manguso N, Gong J, Crystal JS, Paski SC, Hendifar AE, and Tuli R
- Published
- 2021
- Full Text
- View/download PDF
31. Feasibility and efficacy of enteral tube feeding on weight stability, lean body mass, and patient-reported outcomes in pancreatic cancer cachexia.
- Author
-
Gresham G, Placencio-Hickok VR, Lauzon M, Nguyen T, Kim H, Mehta S, Paski S, Pandol SJ, Osipov A, Gong J, Jamil LH, Nissen N, Lo SK, and Hendifar AE
- Subjects
- Aged, Feasibility Studies, Female, Humans, Male, Middle Aged, Patient Reported Outcome Measures, Prospective Studies, Quality of Life, Cachexia etiology, Cachexia therapy, Enteral Nutrition, Pancreatic Neoplasms complications, Pancreatic Neoplasms therapy
- Abstract
Background: Advanced pancreatic ductal adenocarcinoma (PDAC) is characterized by progressive weight loss and nutritional deterioration. This wasting has been linked to poor survival outcomes, alterations in host defenses, decreased functional ability, and diminished health-related quality of life (HRQOL) in pancreatic cancer patients. There are currently no standardized approaches to the management of pancreatic cancer cachexia. This study explores the feasibility and efficacy of enteral tube feeding of a peptide-based formula to improve weight stability and patient-reported outcomes (PROs) in advanced PDAC patients with cachexia., Methods: This was a single-institution, single-arm prospective trial conducted between April 2015 and March 2019. Eligible patients were adults (>18 years) diagnosed with advanced or locally advanced PDAC and cachexia, defined as greater than 5% unexplained weight loss within 6 months from screening. The study intervention included three 28 day cycles of a semi-elemental peptide-based formula, administered through a jejunal or gastrojejunal feeding tube. The primary outcome was weight stability at 3 months (Cycle 3), defined as weight change less than 0.1 kg/baseline BMI unit from baseline. Secondary outcomes included changes in lean body mass, appendicular lean mass, bone mineral density, fat mass, and percent body fat, as measured with a DEXA scan, HRQOL (EORTC QLQC30) and NIH PROMIS PROs assessed at each cycle. Daily activity (steps, distance, active minutes, heart rate, and sleep) were remotely monitored using a wearable activity monitor (Fitbit) over the 3 month study period., Results: Thirty-six patients were screened for eligibility, 31 patients consented onto study and underwent jejunal tube placement, and 16 patients completed treatment: mean age 67 years (SD 9.3), 43.8% male. Among evaluable patients (n = 16), weight stability was achieved in 10 patients (62.5%), thus completing the trial early. Increases in lean body mass (1273.1, SD: 4078, P = 0.01) and appendicular lean mass (0.45, SD: 0.6, P = 0.02) were observed. Statistically significant improvements at Cycle 3 from baseline were also observed for QLQC30 role function [mean difference (MD): 20.1, P = 0.03], appetite (MD: 27.4, P = 0.02), and global health scores (MD: 13.3, P = 0.05) as well as for NIH PROMIS t-scores for depression (MD: -10.4, P = 0.006) and pain interference (MD: -7.5, P = 0.05). Objectively monitored (Fitbit) activity levels increased, although statistical significance was not reached., Conclusions: Our findings suggest that enteral nutrition support may improve weight stability, lean body mass, appendicular lean mass and PROs in PDAC patients with cachexia who completed treatment, representing a subsample of the study population. The feasibility and role of enteral feeding in routine care remain unclear, and larger and randomized controlled trials are warranted., (© 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2021
- Full Text
- View/download PDF
32. Therapeutic targeting of SLC6A8 creatine transporter suppresses colon cancer progression and modulates human creatine levels.
- Author
-
Kurth I, Yamaguchi N, Andreu-Agullo C, Tian HS, Sridhar S, Takeda S, Gonsalves FC, Loo JM, Barlas A, Manova-Todorova K, Busby R, Bendell JC, Strauss J, Fakih M, McRee AJ, Hendifar AE, Rosen LS, Cercek A, Wasserman R, Szarek M, Spector SL, Raza S, Tavazoie MF, and Tavazoie SF
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Creatine metabolism, Creatine pharmacology, Creatine therapeutic use, Humans, Membrane Transport Proteins, Mice, Mice, Nude, Mutation, Nerve Tissue Proteins metabolism, Plasma Membrane Neurotransmitter Transport Proteins genetics, Plasma Membrane Neurotransmitter Transport Proteins pharmacology, Proto-Oncogene Proteins p21(ras) metabolism, Antineoplastic Agents pharmacology, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Colorectal Neoplasms pathology
- Abstract
Colorectal cancer (CRC) is a leading cause of cancer mortality. Creatine metabolism was previously shown to critically regulate colon cancer progression. We report that RGX-202, an oral small-molecule SLC6A8 transporter inhibitor, robustly inhibits creatine import in vitro and in vivo, reduces intracellular phosphocreatine and ATP levels, and induces tumor apoptosis. RGX-202 suppressed CRC growth across KRAS wild-type and KRAS mutant xenograft, syngeneic, and patient-derived xenograft (PDX) tumors. Antitumor efficacy correlated with tumoral expression of creatine kinase B. Combining RGX-202 with 5-fluorouracil or the DHODH inhibitor leflunomide caused regressions of multiple colorectal xenograft and PDX tumors of distinct mutational backgrounds. RGX-202 also perturbed creatine metabolism in patients with metastatic CRC in a phase 1 trial, mirroring pharmacodynamic effects on creatine metabolism observed in mice. This is, to our knowledge, the first demonstration of preclinical and human pharmacodynamic activity for creatine metabolism targeting in oncology, thus revealing a critical therapeutic target.
- Published
- 2021
- Full Text
- View/download PDF
33. Leveraging patient-reported outcomes (PROs) in patients with pancreatic cancer: The Pancreatic Cancer Action Network (PanCAN) online patient registry experience.
- Author
-
Gupta A, Khalid O, Moravek C, Lamkin A, Matrisian LM, Doss S, Denlinger CS, Coveler AL, Weekes CD, Roeland EJ, Hendifar AE, and Nipp RD
- Subjects
- Adult, Advertising, Aged, Aged, 80 and over, Cancer Care Facilities, Feasibility Studies, Female, Humans, Male, Middle Aged, Pancreatic Neoplasms pathology, Pancreatic Neoplasms psychology, Patient Participation, Precision Medicine, Research, United States, Young Adult, Pancreatic Neoplasms therapy, Patient Reported Outcome Measures, Registries statistics & numerical data
- Abstract
Background: The Pancreatic Cancer Action Network (PanCAN) Patient Registry is an online, pancreatic cancer-specific, global registry enabling patients to self-report sociodemographics, disease/management characteristics, and patient-reported outcomes (PROs). We sought to describe the creation, user experience, and research potential of the PanCAN Registry., Methods: We obtained data to describe (1) the creation of the Registry (questionnaire development, marketing efforts, and regulatory considerations); (2) the user experience (user characteristics and interactions with the registry following inception); and (3) the research potential of the registry (comparing PROs and treatment patterns by age [±65 years] and treatment site [community or academic] for users with de novo metastatic disease)., Results: The Registry was conceived as part of PanCAN's strategic plan for a personalized therapy initiative. PanCAN staff and disease expert consultants developed questionnaires hosted on an electronic PRO platform. Users had the option to include their data in research efforts, and the Registry platform received institutional review board approval. From 7/2015 to 12/2020, 2187 patients visited the registry and 1697 (77.6%) completed at least one survey (median age = 64 years [range: 24-90], 47.9% women, 88.7% White, 34.0% metastatic disease). Among patients with metastatic disease (N = 567), 46.0% were ≥65 years old and 67.5% received treatment at community sites. Patients ≥65 years reported feeling less hopeful about the treatment plan (12.4% vs. 24.3%, p = 0.003), and patients treated at community sites reported more frequent treatment breaks of >2 weeks (58.2% vs. 28.1%, p < 0.001)., Conclusions: Our findings demonstrate the feasibility, usability, and research potential of an online PRO registry for patients with cancer. This description of the PanCAN Registry should inform future registry-building efforts to facilitate standardized PRO reporting and provide a valuable research database., Clinical Trial Registration Number: Not applicable., (© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
34. A phase I/II study of rovalpituzumab tesirine in delta-like 3-expressing advanced solid tumors.
- Author
-
Mansfield AS, Hong DS, Hann CL, Farago AF, Beltran H, Waqar SN, Hendifar AE, Anthony LB, Taylor MH, Bryce AH, Tagawa ST, Lewis K, Niu J, Chung CH, Cleary JM, Rossi M, Ludwig C, Valenzuela R, Luo Y, and Aggarwal R
- Abstract
Delta-like protein 3 (DLL3) is highly expressed in solid tumors, including neuroendocrine carcinomas/neuroendocrine tumors (NEC/NET). Rovalpituzumab tesirine (Rova-T) is a DLL3-targeting antibody-drug conjugate. Patients with NECs and other advanced DLL3-expressing tumors were enrolled in this phase I/II study (NCT02709889). The primary endpoint was safety. Two hundred patients were enrolled: 101 with NEC/NET (large-cell NEC, gastroenteropancreatic NEC, neuroendocrine prostate cancer, and other NEC/NET) and 99 with other solid tumors (melanoma, medullary thyroid cancer [MTC], glioblastoma, and other). The recommended phase II dose (RP2D) was 0.3 mg/kg every 6 weeks (q6w) for two cycles. At the RP2D, grade 3/4 adverse events included anemia (17%), thrombocytopenia (15%), and elevated aspartate aminotransferase (8%). Responses were confirmed in 15/145 patients (10%) treated at 0.3 mg/kg, including 9/69 patients (13%) with NEC/NET. Rova-T at 0.3 mg/kg q6w had manageable toxicity, with antitumor activity observed in patients with NEC/NET, melanoma, MTC, and glioblastoma., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
35. Clinical Outcomes Among Patients With Metastatic Pancreatic Ductal Adenocarcinoma Treated With Liposomal Irinotecan.
- Author
-
Yu KH, Hendifar AE, Alese OB, Draper A, Abdelrahim M, Burns E, Khan G, Cockrum P, Bhak RH, Nguyen C, DerSarkissian M, Duh MS, and Bahary N
- Abstract
Background: The NAPOLI-1 trial demonstrated that liposomal irinotecan in combination with fluorouracil (5-FU) and leucovorin (LV) prolonged survival with a manageable safety profile in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) previously treated with gemcitabine-based therapy. Real-world data on clinical outcomes associated with liposomal irinotecan in NAPOLI-1-based regimens is needed to further substantiate this., Methods: This real-world, retrospective chart review study included patients with mPDAC who received NAPOLI-1-based regimens from six academic centers in the United States. Liposomal irinotecan initiation defined the index date. Overall survival (OS) and progression-free survival (PFS) were assessed with Kaplan-Meier methodology., Results: There were 374 patients evaluated; median age was 68 years, and 51% were female. Among 326 patients with baseline ECOG information, approximately 74% had ECOG score <2. Liposomal irinotecan was administered as a doublet with 5-FU in a NAPOLI-1-based regimen in the first line (1L; 16%), 2L (42%), and 3L+ (42%) of the metastatic setting. For patients treated in 1L, 2L, and 3L+, median [95% confidence interval (CI)] OS was 8.0 [5.1, 11.2], 7.3 [5.3, 8.8], and 4.6 [4.0, 5.7] months, and median [95% CI] PFS was 4.2 [2.2, 6.6], 3.0 [2.6, 3.7], and 2.0 [1.7, 2.2] months, respectively., Conclusions: Patients in a real-world setting treated with NAPOLI-1-based liposomal irinotecan doublet regimens at academic centers were older with poorer performance status compared to trial patients yet had similar outcomes and efficacy. Furthermore, liposomal irinotecan was frequently used in the 3L+ setting where no treatment has been approved and provided clinical benefit., Competing Interests: KHY: Research Funding (BMS, Ipsen, Halozyme); Advisory Board (Ipsen). AEH: Consulting or Advisory Role (Novartis, Ipsen, Perthera, Celgene, Abbvie); Research Funding (Ipsen); Travel, Accommodations, Expenses (Halozyme). OBA: Research Funding (Taiho Oncology, Ipsen Pharmaceuticals, GSK, Bristol Myers Squibb, PCI Biotech AS, Calithera Biosciences, Inc., SynCore Biotechnology Co., Ltd., Corcept, Mabspace Biosciences); Consulting/Advisory Role (Exelixis, Conjupro BioTherapeutics, R-Pharm US LLC, Ipsen Pharmaceuticals, Natera, Taiho, Pfizer, QED therapeutics). MA: Advisory Board and Speaker (Ipsen). NB: Consultant (AstraZeneca, Exelixis, BMS, Thermo Fisher). PC is an employee of Ipsen Biopharmaceuticals, Inc. and owns stock/stock options. MSD, MD, RHB, and CN are employees of Analysis Group Inc., which has received consultancy fees from Ipsen Biopharmaceuticals, Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Yu, Hendifar, Alese, Draper, Abdelrahim, Burns, Khan, Cockrum, Bhak, Nguyen, DerSarkissian, Duh and Bahary.)
- Published
- 2021
- Full Text
- View/download PDF
36. Symptom Diaries of Patients with Midgut Neuroendocrine Tumors Treated with 177 Lu-DOTATATE.
- Author
-
Strosberg JR, Srirajaskanthan R, El-Haddad G, Wolin EM, Chasen BR, Kulke MH, Bushnell DL, Caplin ME, Baum RP, Hendifar AE, Öberg K, Ruszniewski P, Santoro P, Broberg P, Leeuwenkamp OR, and Krenning EP
- Abstract
We report the impact of
177 Lu DOTATATE treatment on abdominal pain, diarrhea, and flushing, symptoms that patients with advanced midgut neuroendocrine tumors (NETs) often find burdensome. Methods: All patients enrolled in the international randomized phase 3 Neuroendocrine Tumors Therapy (NETTER-1) trial (177 Lu-DOTATATE plus standard-dose octreotide long-acting repeatable [LAR], n = 117; high-dose octreotide LAR, n = 114) were asked to record the occurrence of predefined symptoms in a daily diary. Change from baseline in symptom scores (mean number of days with a symptom) was analyzed using a mixed model for repeated measures. Results: Patients (intent-to-treat) who received177 Lu-DOTATATE experienced a significantly greater decline from baseline in symptom scores than patients who received high-dose octreotide LAR. For177 Lu-DOTATATE, the mean decline in days with abdominal pain, diarrhea, and flushing was 4.10, 4.55, and 4.52 days per 4 weeks, respectively, compared with 0.99, 1.44, and 2.54 days for high-dose octreotide LAR. The mean differences were 3.11 days (95% confidence interval, 1.35-4.88; P = 0.0007) for abdominal pain, 3.11 days (1.18-5.04; P = 0.0017) for diarrhea, and 1.98 days (0.08-3.88; P = 0.0413) for flushing, favoring177 Lu-DOTATATE. A positive repeated measures correlation was found between diary-recorded symptom scores and questionnaire-recorded pain, diarrhea, and flushing. Conclusion: In addition to efficacy and quality of life benefits, symptom diaries from NETTER-1 demonstrated that treatment with177 Lu DOTATATE was associated with statistically significant reductions in abdominal pain, diarrhea, and flushing, constituting the core symptoms of patients with progressive midgut NETs, compared with high-dose octreotide LAR, supporting a beneficial effect of177 Lu DOTATATE on HRQoL., (Copyright © 2021 by the Society of Nuclear Medicine and Molecular Imaging, Inc.)- Published
- 2021
- Full Text
- View/download PDF
37. Symptom Management in Pancreatic Cancer.
- Author
-
Lee KG, Roy V, Laszlo M, Atkins KM, Lin KJ, Tomassian S, and Hendifar AE
- Subjects
- Adenocarcinoma complications, Adenocarcinoma epidemiology, Adenocarcinoma genetics, Carcinoma, Pancreatic Ductal complications, Carcinoma, Pancreatic Ductal epidemiology, Carcinoma, Pancreatic Ductal genetics, Fatigue complications, Fatigue epidemiology, Fatigue genetics, Fatigue therapy, Humans, Mutation genetics, Pain pathology, Palliative Care, Quality of Life, Adenocarcinoma therapy, Carcinoma, Pancreatic Ductal therapy, Pain epidemiology, Pain Management
- Abstract
Opinion Statement: Despite extensive research that has identified new risk factors, genetic mutations, and therapeutic options, pancreatic ductal adenocarcinoma continues to be a leading cause of cancer related death. Patients with pancreatic cancer, along with their clinicians, must balance realistic hope alongside a life-threatening diagnosis. As the search for treatments to reduce the morbidity and mortality continues, symptom management and quality of life remain the focus of our efforts. In addition to side effects of cancer-directed therapy, patients are at risk for malnutrition, pain, and fatigue. These factors are often overlooked in practice, so a multidisciplinary team is critical in optimizing the care of patients.
- Published
- 2021
- Full Text
- View/download PDF
38. Raynaud's Phenomenon From PD-1 Immune Checkpoint Inhibition.
- Author
-
Kim H, Jones AJ, Labadzhyan A, Placencio-Hickok VR, Wallace DJ, Gong J, and Hendifar AE
- Subjects
- Humans, Programmed Cell Death 1 Receptor, Immune Checkpoint Inhibitors, Raynaud Disease
- Published
- 2020
- Full Text
- View/download PDF
39. Midgut Neuroendocrine Tumors with Liver-only Metastases: Benefit of Primary Tumor Resection.
- Author
-
Gangi A, Manguso N, Gong J, Crystal JS, Paski SC, Hendifar AE, and Tuli R
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Humans, Middle Aged, Retrospective Studies, Survival Rate, Young Adult, Intestinal Neoplasms pathology, Intestinal Neoplasms surgery, Liver Neoplasms secondary, Liver Neoplasms surgery, Neuroendocrine Tumors pathology, Neuroendocrine Tumors surgery
- Abstract
Background: Management of metastatic midgut neuroendocrine tumors (MNET) remains controversial. The benefits of resecting the primary tumor are not clear and advocated only for select patients. This study aimed to determine whether resection of the primary MNET in patients with untreated liver-only metastases has an impact on survival., Methods: This retrospective study reviewed data of the National Cancer Database from 2004 to 2015 for patients with liver-only metastatic MNETs and compared those who received resection of their primary MNET with those who did not. Patient demographics, tumor characteristics, and clinical outcomes were compared between the groups. The primary outcome was overall survival (OS) after adjustment for patient, demographic, and tumor-related factors., Results: The study identified 1952 patients with a median age of 63 years (range, 18-90 years). The median primary tumor size was 2.4 cm (range, 0.1-20 cm). Of these patients, 1295 (66%) underwent resection of the primary tumor and 667 (34%) did not. The patients who underwent resection were younger (median age, 63 vs 65 years; p < 0.001) and had smaller primary tumors (median, 2.3 vs 3.0 cm; p < 0.001). The patients with clinical T1 or T2 tumors were significantly less likely to undergo resection than those with stage T3 or T4 tumors (58.5% vs 89.7%; p < 0.001). The median follow-up period was 43 months (range, 1-83 months). In the entire cohort, 483 deaths occurred, with a 5-year OS of 61%. The 5-year OS rate was 49% for the patients who underwent resection and 66% for those who did not (p < 0.001). When the patients were grouped according to T stage, no OS difference between resection and no resection for stages T1 (p = 0.07) and T2 (p = 0.40) was identified. However, the 5-year OS rate was significantly better for the resected patient cohort with T3 (67.5% vs 37.2%; p < 0.001) or T4 (59.8% vs 21.5%; p < 0.001) tumors., Conclusions: The patients with treatment-naïve liver-only metastatic MNET had improved OS when the primary tumor was resected, particularly those with clinical stage T3 or T4 tumors. These patients may benefit from surgical resection of their primary tumor.
- Published
- 2020
- Full Text
- View/download PDF
40. Randomized Phase III Trial of Pegvorhyaluronidase Alfa With Nab-Paclitaxel Plus Gemcitabine for Patients With Hyaluronan-High Metastatic Pancreatic Adenocarcinoma.
- Author
-
Van Cutsem E, Tempero MA, Sigal D, Oh DY, Fazio N, Macarulla T, Hitre E, Hammel P, Hendifar AE, Bates SE, Li CP, Hingorani SR, de la Fouchardiere C, Kasi A, Heinemann V, Maraveyas A, Bahary N, Layos L, Sahai V, Zheng L, Lacy J, Park JO, Portales F, Oberstein P, Wu W, Chondros D, and Bullock AJ
- Subjects
- Aged, Albumins administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal secondary, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Disease Progression, Double-Blind Method, Fatigue chemically induced, Female, Humans, Hyaluronic Acid metabolism, Hyaluronoglucosaminidase administration & dosage, Hyponatremia chemically induced, Male, Middle Aged, Paclitaxel administration & dosage, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Progression-Free Survival, Response Evaluation Criteria in Solid Tumors, Spasm chemically induced, Survival Rate, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Purpose: To evaluate the efficacy and safety of pegvorhyaluronidase alfa (PEGPH20) plus nab-paclitaxel/gemcitabine (AG) in patients with hyaluronan-high metastatic pancreatic ductal adenocarcinoma (PDA)., Patients and Methods: HALO 109-301 was a phase III, randomized, double-blind, placebo-controlled study. Patients ≥ 18 years of age with untreated, metastatic, hyaluronan-high PDA were randomly assigned 2:1 to PEGPH20 plus AG or placebo plus AG. Treatment was administered intravenously in 4-week cycles (3 weeks on, 1 week off) until progression or intolerable adverse events: PEGPH20 3.0 µg/kg twice per week for cycle 1 and once per week thereafter; nab-paclitaxel 125 mg/m
2 once per week; and gemcitabine 1,000 mg/m2 once per week. The primary end point was overall survival (OS); secondary end points included progression-free survival (PFS), objective response rate (ORR), and safety. Response was independently assessed per RECIST v1.1., Results: At data cutoff, 494 patients were randomly assigned, with 492 (327 for PEGPH20 and 165 for placebo) included in intention-to-treat analyses. Baseline characteristics were balanced for PEGPH20 plus AG versus placebo plus AG. There were 330 deaths, with a median OS of 11.2 months for PEGPH20 plus AG versus 11.5 months for placebo plus AG (hazard ratio [HR], 1.00; 95% CI, 0.80 to 1.27; P = .97); median PFS was 7.1 months versus 7.1 months (HR, 0.97 [95% CI, 0.75 to 1.26]); ORR was 47% versus 36% (ORR ratio, 1.29 [95% CI, 1.03 to 1.63]). Grade ≥ 3 adverse events with a ≥ 2% higher rate with PEGPH20 plus AG than with placebo plus AG included fatigue (16.0% v 9.6%), muscle spasms (6.5% v 0.6%), and hyponatremia (8.0% v 3.8%)., Conclusion: The addition of PEGPH20 to AG increased the ORR but did not improve OS or PFS. The safety profile of PEGPH20 plus AG was consistent with that found in previous studies. These results do not support additional development of PEGPH20 in metastatic PDA.- Published
- 2020
- Full Text
- View/download PDF
41. Remote Oncology Care: Review of Current Technology and Future Directions.
- Author
-
McGregor BA, Vidal GA, Shah SA, Mitchell JD, and Hendifar AE
- Abstract
Cancer patients frequently develop tumor and treatment-related complications, leading to diminished quality of life, shortened survival, and overutilization of emergency department and hospital services. Outpatient oncology treatment has potential to leave cancer patients unmonitored for long periods while at risk of clinical deterioration which has been exaggerated during the COVID19 pandemic. Visits to cancer clinics and hospitals risk exposing immunocompromised patients to infectious complications. Remote patient reported outcomes monitoring systems have been developed for use in cancer treatment, showing benefits in economic and survival outcomes. While advanced devices such as pulmonary artery pressure monitors and implantable loop recorders have proven benefits in cardiovascular care, similar options do not exist for oncology. Here we review the current literature around remote patient monitoring in cancer care and propose the use of reliable devices for capturing and reporting patient symptoms and physiology., Competing Interests: Dr. Mitchell is a named inventor on six patent applications related to this work., (Copyright © 2020, McGregor et al.)
- Published
- 2020
- Full Text
- View/download PDF
42. Priority Rankings of Patient-Reported Outcomes for Pancreatic Ductal Adenocarcinoma: A Comparison of Patient and Physician Perspectives.
- Author
-
Guan M, Gresham G, Shinde A, Lapite I, Gong J, Placencio-Hickok VR, Forrest CB, and Hendifar AE
- Subjects
- Activities of Daily Living, Aged, Female, Humans, Male, Physicians, Carcinoma, Pancreatic Ductal therapy, Pancreatic Neoplasms therapy, Patient Reported Outcome Measures
- Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is associated with high symptom burden. However, treatment decisions currently depend heavily on physician interpretation of clinical parameters and may not consider patients' health preferences. The NIH Patient-Reported Outcomes Measurement Information System (PROMIS) initiative standardized a set of patient-reported outcomes for use in chronic diseases. This study identifies preference rankings among patients with PDAC and physicians for PROMIS domains and compares the priorities of patients and their providers., Methods: We condensed the 96 NIH PROMIS adult domains into 31 domains and created a Maximum Difference Scaling questionnaire. Domain preference scores were generated from the responses of patients with PDAC and physicians, which were compared using Maximum Difference Scaling software across demographic and clinical variables., Results: Participants included 135 patients with PDAC (53% male; median age, 68 years) and 54 physicians (76% male; median years of experience, 10). Patients selected physical functioning (PF) as their top priority, whereas physicians identified pain as most important. PF, ability to perform activities of daily living, and symptom management were within the top 5 domains for both patients and physicians, and varied only slightly across age, sex, and ethnicity. However, several domains were ranked significantly higher by patients than by physicians, including but not limited to PF; ability to do things for yourself, family, and friends; ability to interact with others to obtain help; and sleep quality. Physicians ranked pain, anxiety, and depression higher than patients did., Conclusions: Our findings suggest that patients with PDAC value PF and engaging in daily and social activities the most, whereas physicians prioritize symptoms such as pain. Patient-reported outcomes need to become more integrated into PDAC care and research to better identify unmet patient needs, inform treatment decisions, and develop therapies that address outcomes valued by patients.
- Published
- 2020
- Full Text
- View/download PDF
43. Novel ALK Fusion, PPFIBP1-ALK , in Pancreatic Ductal Adenocarcinoma Responsive to Alectinib and Lorlatinib.
- Author
-
Gower A, Golestany B, Gong J, Singhi AD, and Hendifar AE
- Abstract
Competing Interests: The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/authors/author-center. Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments). Jun GongHonoraria: Amgen, Astellas Pharma, Clinical Congress Consultants, QED Therapeutics, Exelixis, Elsevier Consulting or Advisory Role: Amgen, Astellas Pharma, Clinical Congress Consultants, QED Therapeutics, Exelixis, ElsevierAatur D. SinghiHonoraria: Foundation MedicineAndrew Eugene HendifarConsulting or Advisory Role: Novartis, Ipsen, Perthera, Celgene, AbbVie Research Funding: Ipsen Travel, Accommodations, Expenses: Halozyme No other potential conflicts of interest were reported.
- Published
- 2020
- Full Text
- View/download PDF
44. Dual Checkpoint Blockade in a Neuroendocrine Carcinoma With Dual PD-L1/PD-L2 Amplification and High Tumor Mutational Burden.
- Author
-
Gong J, Patel S, Adashek JJ, Frishberg D, Guan M, Placencio-Hickok VR, Gangi A, Gresham G, Tuli R, Chae YK, Kurzrock R, and Hendifar AE
- Abstract
Competing Interests: The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center. Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments). Jun GongHonoraria: Amgen, Astellas Pharma, Clinical Congress Consultants Consulting or Advisory Role: Amgen, Astellas Pharma, Clinical Congress ConsultantsSandip PatelConsulting or Advisory Role: Eli Lilly, Novartis, Bristol-Myers Squibb, AstraZeneca/MedImmune, Nektar, Compugen, Illumina Speakers' Bureau: Merck, Boehringer Ingelheim Research Funding: Bristol-Myers Squibb (Inst), Pfizer (Inst), Roche (Inst), Amgen (Inst), AstraZeneca/MedImmune (Inst), Fate (Inst), Merck (Inst)David FrishbergStock and Other Ownership Interests: Amgen (I) Consulting or Advisory Role: Ultragenyx (I), Replimmune (I) Travel, Accommodations, Expenses: Ultragenyx (I), Replimmune (I)Veronica R. Placencio-HickokPatents, Royalties, Other Intellectual Property: Patent 17814046.3-1111 PCT/US2017037558. Sensitization of tumors to therapies through endoglin antagonism (Inst)Richard TuliConsulting or Advisory Role: AstraZeneca Research Funding: AstraZeneca, AbbVie Patents, Royalties, Other Intellectual Property: Patent pending: Methods of Treating Gastrointestinal Malignancies, Application No. 16/256,840 filed: January 24, 2019, inventor (Inst)Young K. ChaeConsulting or Advisory Role: Foundation Medicine, Boehringer Ingelheim, Biodesix, Counsyl, AstraZeneca, Guardant Health, Takeda, Roche, Immuneoncia, Hanmi, Eli Lilly, Tempus Speakers' Bureau: Roche, Merck, AstraZeneca, Eli Lilly Research Funding: AbbVie, Bristol-Myers Squibb, Lexent Bio, Freenome, Biodesix Travel, Accommodations, Expenses: HanmiRazelle KurzrockLeadership: CureMatch, CureMetrix Stock and Other Ownership Interests: CureMatch, IDbyDNA, Soluventis Honoraria: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed Therapeutics, Advanced Therapeutics, LEK, AACR, Chugai Pharma USA, Wiley Consulting or Advisory Role: Actuate Therapeutics, Loxo, XBiotech, Neo-Med, Roche, Gaido, Soluventis, Pfizer, Merck Speakers' Bureau: Roche Research Funding: Guardant Health (Inst), Sequenom (Inst), Merck Serono (Inst), Genentech (Inst), Pfizer (Inst), Foundation Medicine (Inst), Incyte (Inst), Konica Minolta (Inst), Grifols (Inst), OmniSeq (Inst), Debiopharm Group (Inst), Boehringer Ingelheim (Inst) Travel, Accommodations, Expenses: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed Therapeutics, Advanced Therapeutics, LEK, AACR, Chugai Pharma USA, WileyAndrew E. HendifarConsulting or Advisory Role: Novartis, Ipsen, Perthera, Celgene, AbbVie Research Funding: Ipsen Travel, Accommodations, Expenses: Halozyme No other potential conflicts of interest were reported.
- Published
- 2020
- Full Text
- View/download PDF
45. Landscape of Health-Related Quality of Life in Patients With Early-Stage Pancreatic Cancer Receiving Adjuvant or Neoadjuvant Chemotherapy: A Systematic Literature Review.
- Author
-
Macarulla T, Hendifar AE, Li CP, Reni M, Riess H, Tempero MA, Dueck AC, Botteman MF, Deshpande CG, Lucas EJ, and Oh DY
- Subjects
- Aged, Chemotherapy, Adjuvant, Female, Humans, Male, Middle Aged, Minimal Clinically Important Difference, Neoplasm Staging, Pancreatic Neoplasms diagnosis, Predictive Value of Tests, Time Factors, Treatment Outcome, Health Status Indicators, Neoadjuvant Therapy adverse effects, Pancreatectomy adverse effects, Pancreatic Neoplasms therapy, Patient Reported Outcome Measures, Quality of Life
- Abstract
Objectives: Pancreatic resection is associated with postoperative morbidity and reduced quality of life (QoL). A systematic literature review was conducted to understand the patient-reported outcome measure (PROM) landscape in early-stage pancreatic cancer (PC)., Methods: Databases/registries (through January 24, 2019) and conference abstracts (2014-2017) were searched. Study quality was assessed using the Newcastle-Ottawa Scale/Cochrane risk-of-bias tool. Searches were for general (resectable PC, adjuvant/neoadjuvant, QoL) and supplemental studies (resectable PC, European Organisation for Research and Treatment of Cancer QoL Questionnaire [QLQ] - Pancreatic Cancer [PAN26])., Results: Of 750 studies identified, 39 (general, 22; supplemental, 17) were eligible: 32 used QLQ Core 30 (C30) and/or QLQ-PAN26, and 15 used other PROMs. Baseline QLQ-C30 global health status/QoL scores in early-stage PC were similar to all-stage PC reference values but lower than all-stage-all-cancer values. The QoL declined after surgery, recovered to baseline in 3 to 6 months, and then generally stabilized. A minimally important difference (MID) of 10 was commonly used for QLQ-C30 but was not established for QLQ-PAN26., Conclusions: In early-stage PC, QLQ-C30 and QLQ-PAN26 are the most commonly used PROMs. Baseline QLQ-C30 global health status/QoL scores suggested a high humanistic burden. Immediately after surgery, QoL declined but seemed stable over the longer term. The QLQ-C30 MID may elucidate the clinical impact of treatment on QoL; MID for QLQ-PAN26 needs to be established.
- Published
- 2020
- Full Text
- View/download PDF
46. Multiparametric Mapping Magnetic Resonance Imaging of Pancreatic Disease.
- Author
-
Wang L, Gaddam S, Wang N, Xie Y, Deng Z, Zhou Z, Fan Z, Jiang T, Christodoulou AG, Han F, Lo SK, Wachsman AM, Hendifar AE, Pandol SJ, and Li D
- Abstract
Background: Current magnetic resonance imaging (MRI) of pancreatic disease is qualitative in nature. Quantitative imaging offers several advantages, including increased reproducibility and sensitivity to detect mild or diffuse disease. The role of multiparametric mapping MRI in characterizing various tissue types in pancreatic disease such as chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC) has rarely been evaluated., Purpose: To evaluate the feasibility of multiparametric mapping [T1, T2, and apparent diffusion coefficient (ADC)] in defining tissue characteristics that occur in CP and PDAC to improve disease diagnosis., Materials and Methods: Pancreatic MRI was performed in 17 patients with PDAC undergoing therapy, 7 patients with CP, and 29 healthy volunteers with no pancreatic disease. T1 modified Look-Locker Inversion Recovery (T1 MOLLI), T2-prepared gradient-echo, and multi-slice single-shot echo-planar diffusion weighted imaging (SS-EPI DWI) sequences were used for data acquisition. Regions of interest (ROIs) of pancreas in PDAC, CP, and control subjects were outlined by an experienced radiologist. One-way analysis of variance (ANOVA) was used to compare the difference between groups and regions of the pancreas, and Tukey tests were used for multiple comparison testing within groups. Receiver operator characteristic (ROC) curves were analyzed, and the areas under the curves (AUCs) were calculated using single parameter and combined parameters, respectively., Results: T1, T2, and ADC values of the entire pancreas among PDAC, CP, and control subjects; and between upstream and downstream portions of the pancreas in PDAC patients were all significantly different ( p < 0.05). The AUC values were 0.90 for T1, 0.55 for T2, and 0.71 for ADC for independent prediction of PDAC. By combining T1, T2, and ADC, the AUC value was 0.94 (sensitivity 91.54%, specificity 85.81%, 95% CI: 0.92-0.96), which yielded higher accuracy than any one parameter only ( p < 0.001)., Conclusion: Multiparametric mapping MRI is feasible for the evaluation of the differences between PDAC, CP, and normal pancreas tissues. The combination of multiple parameters of T1, T2, and ADC provides a higher accuracy than any single parameter alone in tissue characterization of the pancreas., (Copyright © 2020 Wang, Gaddam, Wang, Xie, Deng, Zhou, Fan, Jiang, Christodoulou, Han, Lo, Wachsman, Hendifar, Pandol and Li.)
- Published
- 2020
- Full Text
- View/download PDF
47. An open-label, single-arm pilot study of EUS-guided brachytherapy with phosphorus-32 microparticles in combination with gemcitabine +/- nab-paclitaxel in unresectable locally advanced pancreatic cancer (OncoPaC-1): Technical details and study protocol.
- Author
-
Bhutani MS, Klapman JB, Tuli R, El-Haddad G, Hoffe S, Wong FCL, Chasen B, Fogelman DR, Lo SK, Nissen NN, Hendifar AE, Varadhachary G, Katz MHG, Erwin WD, Koay EJ, Tamm EP, Singh BS, Mehta R, Wolff RA, Soman A, Cazacu IM, and Herman JM
- Abstract
Current treatment options for patients with unresectable locally advanced pancreatic cancer (LAPC) include chemotherapy alone or followed by chemoradiation or stereotactic body radiotherapy. However, the prognosis for these patients remains poor, with a median overall survival <12 months. Therefore, novel treatment options are needed. Currently, there is no brachytherapy device approved for pancreatic cancer treatment. Hereby, we present the protocol of a prospective, multicenter, interventional, open-label, single-arm pilot study (OncoPac-1, Clinicaltrial.gov-NCT03076216) aiming to determine the safety and efficacy of Phosphorus-32 when implanted directly into pancreatic tumors using EUS guidance, for patients with unresectable LAPC undergoing chemotherapy (gemcitabine ± nab-paclitaxel)., Competing Interests: None
- Published
- 2020
- Full Text
- View/download PDF
48. Efficacy of PD-1 Blockade in Refractory Microsatellite-Stable Colorectal Cancer With High Tumor Mutation Burden.
- Author
-
Gong J, Robertson MD, Kim E, Fakih M, Schrock AB, Tam KW, Burugapalli B, Monjazeb AM, Hendifar AE, Hitchins M, Klempner SJ, and Cho M
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma pathology, Antineoplastic Agents, Immunological therapeutic use, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, DNA Mutational Analysis, DNA-Binding Proteins genetics, High-Throughput Nucleotide Sequencing, Humans, Male, Middle Aged, Prognosis, Transcription Factors genetics, Transforming Growth Factor beta1 genetics, Antibodies, Monoclonal, Humanized therapeutic use, Biomarkers, Tumor genetics, Colorectal Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Microsatellite Instability, Mutation, Programmed Cell Death 1 Receptor antagonists & inhibitors
- Published
- 2019
- Full Text
- View/download PDF
49. Multiplatform profiling of pancreatic neuroendocrine tumors: Correlative analyses of clinicopathologic factors and identification of co-occurring pathogenic alterations.
- Author
-
Gong J, Blais EM, Bender JR, Guan M, Placencio-Hickok V, Petricoin EF, Pishvaian MJ, Gregory G, Tuli R, and Hendifar AE
- Abstract
Background: Multi-omic profiling of pancreatic neuroendocrine tumors (PanNETs) was performed to correlate genomic, proteomic, and molecular pathway alterations with clinicopathologic factors and identify novel co-occurring pathogenic alterations of potential clinical relevance to PanNET management., Methods: PanNETs referred to Perthera, Inc. having undergone molecular profiling for precision matched therapeutic purposes were screened. Correlative analyses were performed using Fisher's exact test across individual pathogenic alterations or altered molecular pathways and clinicopathologic variables. Associations were visualized by hierarchical clustering. Prognostic associations with overall survival (OS) were identified using Cox regression for pathogenic alterations and pathway-level alterations. Hazard ratios (HR) and odds ratios (OR) were reported with 95% confidence intervals (CI)., Results: From 12/2014-1/2019, 46 patients with predominantly locally advanced and metastatic PanNETs were included. MEN1 alterations by next-generation sequencing (NGS) were less associated with having high-grade PanNETs and metastatic disease at diagnosis ( p ≤ 0.05). Genomic alterations associated with increased replicative stress (primarily driven by RB1 and TP53 ) correlated with higher grade (OR 6.87 [95% CI: 1.57-35.18], p = 0.0043) and worse OS (HR 13.62 [95% CI: 1.51-122.5], p = 0.0198). Other significant associations included: ERCC1 protein expression with DAXX or MEN1 alterations (NGS), PTEN (NGS) with ARID1A or TP53 alterations (NGS), and history of diabetes coincided with cell cycle pathway alterations but was mutually exclusive with replicative stress pathway alterations., Conclusions: We identified several molecular signatures of potential clinical significance for therapeutic targeting and prognostication in PanNETs warranting prospective validation. Our findings are hypothesis generating and can inform larger molecular profiling efforts in PanNETs., Competing Interests: CONFLICTS OF INTEREST E.M.B., E.F.P., M.J.P., and G.G. hold ownership interest (including patents) in Perthera. E.F.P., M.J.P., and A.E.H. are consultant/advisory board members for Perthera. No conflicts of interest were reported by J.G., J.R.B., M.G., V.P., and R.T.
- Published
- 2019
- Full Text
- View/download PDF
50. A virtual molecular tumor board to improve efficiency and scalability of delivering precision oncology to physicians and their patients.
- Author
-
Pishvaian MJ, Blais EM, Bender RJ, Rao S, Boca SM, Chung V, Hendifar AE, Mikhail S, Sohal DPS, Pohlmann PR, Moore KN, He K, Monk BJ, Coleman RL, Herzog TJ, Halverson DD, DeArbeloa P, Petricoin EF 3rd, and Madhavan S
- Abstract
Objectives: Scalable informatics solutions that provide molecularly tailored treatment recommendations to clinicians are needed to streamline precision oncology in care settings., Materials and Methods: We developed a cloud-based virtual molecular tumor board (VMTB) platform that included a knowledgebase, scoring model, rules engine, an asynchronous virtual chat room and a reporting tool that generated a treatment plan for each of the 1725 patients based on their molecular profile, previous treatment history, structured trial eligibility criteria, clinically relevant cancer gene-variant assertions, biomarker-treatment associations, and current treatment guidelines. The VMTB systematically allows clinician users to combine expert-curated data and structured data from clinical charts along with molecular testing data to develop consensus on treatments, especially those that require off-label and clinical trial considerations., Results: The VMTB was used as part of the cancer care process for a focused subset of 1725 patients referred by advocacy organizations wherein resultant personalized reports were successfully delivered to treating oncologists. Median turnaround time from data receipt to report delivery decreased from 14 days to 4 days over 4 years while the volume of cases increased nearly 2-fold each year. Using a novel scoring model for ranking therapy options, oncologists chose to implement the VMTB-derived therapies over others, except when pursuing immunotherapy options without molecular support., Discussion: VMTBs will play an increasingly critical role in precision oncology as the compendium of biomarkers and associated therapy options available to a patient continues to expand., Conclusion: Further development of such clinical augmentation tools that systematically combine patient-derived molecular data, real-world evidence from electronic health records and expert curated knowledgebases on biomarkers with computational tools for ranking best treatments can support care pathways at point of care., (© The Author(s) 2019. Published by Oxford University Press on behalf of the American Medical Informatics Association.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.