551
|
Valdera FA, O'Shea AE, Smolinsky TR, Carpenter EL, Adams AA, McCarthy PM, Tiwari A, Chick RC, Kemp-Bohan PM, Van Decar S, Thomas KK, Bader JO, Peoples GE, Clifton GT, Stojadinovic A, Nelson DW, Vreeland TJ. Predictors and benefits of multiagent chemotherapy for pancreatic adenocarcinoma: Timing matters. J Surg Oncol 2024; 129:244-253. [PMID: 37800378 DOI: 10.1002/jso.27466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/11/2023] [Accepted: 09/17/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Adjuvant (A) multiagent chemotherapy (MC) is the standard of care for patients with pancreatic adenocarcinoma (PDAC). Tolerating MC following a morbid operation may be difficult, thus neoadjuvant (NA) treatment is preferable. This study examined how the timing of chemotherapy was related to the regimen given and ultimately the overall survival (OS). METHODS The National Cancer Database was queried from 2006 to 2017 for nonmetastatic PDAC patients who underwent surgical resection and received MC or single-agent chemotherapy (SC) pre- or postresection. Predictors of receiving MC were determined using multivariable logistic regression. Five-year OS was evaluated using the Kaplan-Meier and Cox proportional hazards model. RESULTS A total of 12,440 patients (NA SC, n = 663; NA MC, n = 2313; A SC, n = 6152; A MC, n = 3312) were included. MC utilization increased from 2006-2010 to 2011-2017 (33.1%-49.7%; odds ratio [OR]: 0.59; p < 0.001). Younger age, fewer comorbidities, higher clinical stage, and larger tumor size were all associated with receipt of MC (all p < 0.001), but NA treatment was the greatest predictor (OR 5.18; 95% confidence interval [CI]: 4.63-5.80; p < 0.001). MC was associated with increased median 5-year OS (26.0 vs. 23.9 months; hazard ratio [HR]: 0.92; 95% CI: 0.88-0.96) and NA MC was associated with the highest survival (28.2 months) compared to NA SC (23.3 months), A SC (24.0 months), and A MC (24.6 months; p < 0.001). CONCLUSION Use and timing of MC contribute to OS in PDAC with an improved 5-year OS compared to SC. The greatest predictor of receiving MC was being given as NA therapy and the greatest survival benefit was the NA MC subgroup. Randomized studies evaluating the timing of effective MC in PDAC are needed.
Collapse
Affiliation(s)
- Franklin A Valdera
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | - Anne E O'Shea
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | - Todd R Smolinsky
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | | | - Alexandra A Adams
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | - Patrick M McCarthy
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | - Ankur Tiwari
- Department of Surgery, University of Texas San Antonio Health Science Center, San Antonio, Texas, USA
| | - Robert C Chick
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | | | - Spencer Van Decar
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | - Katryna K Thomas
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | | | | | - Guy T Clifton
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| | | | - Daniel W Nelson
- Department of Surgery, William Beaumont Army Medical Center, El Paso, Texas, USA
| | - Timothy J Vreeland
- Department of Surgery, Brooke Army Medical Center, Ft. Sam Houston, Texas, USA
| |
Collapse
|
552
|
Tamaki A, Kato T, Sakurai Y, Sato K, Adachi K, Tadehara M, Kogami T, Matsushita M, Hoshino A, Sanoyama I, Numata Y, Umezawa A, Ichinoe M, Ichihara M, Kusano C, Murakumo Y. REV7 is involved in outcomes of platinum-based chemotherapy in pancreatic cancer by controlling the DNA damage response. Cancer Sci 2024; 115:660-671. [PMID: 38130032 PMCID: PMC10859597 DOI: 10.1111/cas.16044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
REV7 is a multifunctional protein implicated in various biological processes, including DNA damage response. REV7 expression in human cancer cells affects their sensitivity to DNA-damaging agents. In the present study, we investigated the significance of REV7 in pancreatic ductal adenocarcinoma (PDAC). REV7 expression was immunohistochemically examined in 92 resected PDAC specimens and 60 endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNAB) specimens of unresectable PDAC treated with platinum-based chemotherapy, and its association with clinicopathologic features was analyzed. Although REV7 expression was not significantly associated with the progression of primary tumors (T-factor and Stage) in either resected or unresectable PDAC, decreased levels of REV7 expression in EUS-FNAB specimens of unresectable PDAC were significantly associated with better outcomes of platinum-based chemotherapy and a favorable prognosis. REV7-deficient PDAC cell lines showed suppressed cell growth and enhanced sensitivity to cisplatin in vitro. Tumor-bearing mice generated using REV7-deficient PDAC cell lines also showed enhanced sensitivity to cisplatin in vivo. RNA sequencing analysis using WT and REV7-deficient PDAC cell lines revealed that REV7 inactivation promoted the downregulation of genes involved in the DNA repair and the upregulation of genes involved in apoptosis. Our results indicate that decreased expression of REV7 is associated with better outcomes of platinum-based chemotherapy in PDAC by suppressing the DNA damage response. It is also suggested that REV7 is a useful biomarker for predicting the outcome of platinum-based chemotherapy and the prognosis of unresectable PDAC and is a potential target for PDAC treatment.
Collapse
Affiliation(s)
- Akihiro Tamaki
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Takuya Kato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yasutaka Sakurai
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Keita Sato
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Kai Adachi
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masayoshi Tadehara
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Taro Kogami
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Masahiro Matsushita
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Akiyoshi Hoshino
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Itaru Sanoyama
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiko Numata
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Atsuko Umezawa
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masaaki Ichinoe
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| | - Masatoshi Ichihara
- Department of Biomedical Sciences, College of Life and Health SciencesChubu UniversityKasugaiJapan
| | - Chika Kusano
- Department of GastroenterologyKitasato University School of MedicineSagamiharaJapan
| | - Yoshiki Murakumo
- Department of PathologyKitasato University School of MedicineSagamiharaJapan
| |
Collapse
|
553
|
Sugawara T, Rodriguez Franco S, Sherman S, Torphy RJ, Colborn K, Franklin O, Ishida J, Grandi S, Al-Musawi MH, Gleisner A, Schulick RD, Del Chiaro M. Neoadjuvant Chemotherapy Versus Upfront Surgery for Resectable Pancreatic Adenocarcinoma: An Updated Nationwide Study. Ann Surg 2024; 279:331-339. [PMID: 37226812 DOI: 10.1097/sla.0000000000005925] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
OBJECTIVE The objective of this study was to assess the association of survival with neoadjuvant chemotherapy (NAC) in resectable pancreatic adenocarcinoma (PDAC). BACKGROUND The early control of potential micrometastases and patient selection using NAC has been advocated for patients with PDAC. However, the role of NAC for resectable PDAC remains unclear. METHODS Patients with clinical T1 and T2 PDAC were identified in the National Cancer Database from 2010 to 2017. Kaplan-Meier estimates, and Cox regression models were used to compare survival. To address immortal time bias, landmark analysis was performed. Interactions between preoperative factors and NAC were investigated in subgroup analyses. A propensity score analysis was performed to compare survival between multiagent NAC and upfront surgery. RESULTS In total, 4041 patients were treated with upfront surgery and 1,175 patients were treated with NAC (79.4% multiagent NAC, 20.6% single-agent NAC). Using a landmark time of 6 months after diagnosis, patients treated with multiagent NAC had longer median overall survival compared with upfront surgery and single-agent NAC. (35.8 vs 27.1 vs 27.4 mo). Multiagent NAC was associated with lower mortality rates compared with upfront surgery (adjusted hazard ratio, 0.77; 95% CI, 0.70-0.85), whereas single-agent NAC was not. The association of survival with multiagent NAC were consistent in analyses using the matched data sets. Interaction analysis revealed that the association between multiagent NAC and a lower mortality rate did not significantly differ across age, facility type, tumor location, CA 19-9 levels, and clinical T/N stages. CONCLUSIONS The findings suggest that multiagent NAC followed by resection is associated with improved survival compared with upfront surgery.
Collapse
Affiliation(s)
- Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Salvador Rodriguez Franco
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
| | - Samantha Sherman
- Department of Surgery, Parkview Hospital Randallia, Fort Wayne, IN
| | - Robert J Torphy
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO
| | - Kathryn Colborn
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO
- Surgical Outcomes and Applied Research Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
| | - Oskar Franklin
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Jun Ishida
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
| | - Samuele Grandi
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
| | | | - Ana Gleisner
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO
| | - Richard D Schulick
- Surgical Outcomes and Applied Research Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
554
|
Yun WG, Han Y, Cho YJ, Jung HS, Lee M, Kwon W, Jang JY. In Neoadjuvant FOLFIRINOX Chemotherapy for Pancreatic Ductal Adenocarcinoma, Which Response is the More Reliable Indicator for Prognosis, Radiologic or Biochemical? Ann Surg Oncol 2024; 31:1336-1346. [PMID: 37991581 DOI: 10.1245/s10434-023-14532-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/18/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND In this era of increasing neoadjuvant chemotherapy, methods for evaluating responses to neoadjuvant chemotherapy are still diverse among institutions. Additionally, the efficacy of adjuvant chemotherapy for patients undergoing neoadjuvant chemotherapy remains unclear. Therefore, this retrospective study was performed to evaluate the effectiveness of methods for assessing response to neoadjuvant chemotherapy and the need for adjuvant chemotherapy in treating patients with non-metastatic pancreatic ductal adenocarcinoma. METHODS The study identified 150 patients who underwent neoadjuvant FOLFIRINOX chemotherapy followed by curative-intent pancreatectomy. The patients were stratified by biochemical response based on the normalization of carbohydrate antigen 19-9 and by radiologic response based on size change at imaging. RESULTS The patients were classified into the following three groups based on their response to neoadjuvant chemotherapy and prognosis: biochemical responders (BR+), radiology-only responders (BR-/RR+), and non-responders (BR-/RR-). The 3-year overall survival rate was higher for BR+ (71.0%) than for BR-/RR+ (53.6%) or BR-/RR- (33.1%) (P < 0.001). Response to neoadjuvant chemotherapy also was identified as a significant risk factor for recurrence in a comparison between BR-/RR+ and BR+ (hazard ratio [HR], 2.15; 95% confidence interval [CI] 1.19-3.88; P = 0.011) and BR-/RR- (HR, 3.82; 95% CI 2.41-6.08; P < 0.001). Additionally, regardless of the response to neoadjuvant chemotherapy, patients who completed adjuvant chemotherapy had a significantly higher 3-year overall survival rate than those who did not. CONCLUSIONS This response evaluation criterion for neoadjuvant chemotherapy is feasible and can significantly predict prognosis. Additionally, completion of adjuvant chemotherapy could be helpful to patients who undergo neoadjuvant chemotherapy regardless of their response to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jae Cho
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
555
|
Scianò F, Terrana F, Pecoraro C, Parrino B, Cascioferro S, Diana P, Giovannetti E, Carbone D. Exploring the therapeutic potential of focal adhesion kinase inhibition in overcoming chemoresistance in pancreatic ductal adenocarcinoma. Future Med Chem 2024; 16:271-289. [PMID: 38269431 DOI: 10.4155/fmc-2023-0234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 01/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related deaths worldwide. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase often overexpressed in PDAC. FAK has been linked to cell migration, survival, proliferation, angiogenesis and adhesion. This review first highlights the chemoresistant nature of PDAC. Second, the role of FAK in PDAC cancer progression and resistance is carefully described. Additionally, it discusses recent developments of FAK inhibitors as valuable drugs in the treatment of PDAC, with a focus on diamine-substituted-2,4-pyrimidine-based compounds, which represent the most potent class of FAK inhibitors in clinical trials for the treatment of PDAC disease. To conclude, relevant computational studies performed on FAK inhibitors are reported to highlight the key structural features required for interaction with the protein, with the aim of optimizing this novel targeted therapy.
Collapse
Affiliation(s)
- Fabio Scianò
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Francesca Terrana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Camilla Pecoraro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Barbara Parrino
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Stella Cascioferro
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Patrizia Diana
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, San Giuliano Terme, Pisa, 56017, Italy
| | - Daniela Carbone
- Department of Biological, Chemical & Pharmaceutical Sciences & Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| |
Collapse
|
556
|
Nagashima S, Kobayashi S, Tsunoda S, Yamachika Y, Tozuka Y, Fukushima T, Morimoto M, Ueno M, Furuse J, Maeda S. Liposomal irinotecan plus fluorouracil/leucovorin in older patients with advanced pancreatic cancer: a single-center retrospective study. Int J Clin Oncol 2024; 29:188-194. [PMID: 37991558 DOI: 10.1007/s10147-023-02432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND The global phase 3 NAPOLI -1 trial of patients with pancreatic ductal adenocarcinoma (PDAC) demonstrated an overall survival (OS) benefit from using liposomal irinotecan and 5-fluorouracil/leucovorin (nal-IRI + 5-FU/LV) after treatment with gemcitabine (GEM) compared to 5-FU/LV alone. However, the efficacy and safety of this regimen in older patients are not well studied. METHODS We conducted a single-center retrospective study to compare the therapeutic efficacy of nal-IRI + 5-FU/LV between older and younger patients with cutoff ages of 70 and 75 years, respectively. We included patients with a prior history of one or more GEM-based regimens for locally advanced or metastatic PDAC and were treated with nal-IRI + 5-FU/LV. RESULTS Of the 115 patients, 54 (47.0%) and 24 (20.9%) were aged ≥ 70 and ≥ 75 years, respectively. The median OS and progression-free survival (PFS) of the entire cohort were 8.5 and 3.6 months, respectively. No significant differences were observed in OS and PFS hazard ratios using age cutoffs of 70 (P = 0.90 and 0.99, respectively) and 75 (P = 0.90 and 0.76, respectively) years. Additionally, no significant differences were found in the incidence of treatment-related adverse events (trAEs) between patients aged ≥ 70 and < 70 years or those aged ≥ 75 and < 75 years. Other than hematological toxicity, no trAEs higher than Grade 4 were observed in either age group. CONCLUSION The efficacy and safety of nal-IRI + 5-FU/LV for patients with PDAC are not significantly different for those aged ≥ 70 years compared to younger patients.
Collapse
Affiliation(s)
- Shuhei Nagashima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan.
| | - Shotaro Tsunoda
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Yui Yamachika
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Yuichiro Tozuka
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Manabu Morimoto
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-Ku, Yokohama, 241-8515, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
557
|
Kim S, Leem G, Choi J, Koh Y, Lee S, Nam SH, Kim JS, Park CH, Hwang HK, Min KI, Jo JH, Lee HS, Chung MJ, Park JY, Park SW, Song SY, Shin EC, Kang CM, Bang S, Park JE. Integrative analysis of spatial and single-cell transcriptome data from human pancreatic cancer reveals an intermediate cancer cell population associated with poor prognosis. Genome Med 2024; 16:20. [PMID: 38297291 PMCID: PMC10832111 DOI: 10.1186/s13073-024-01287-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Recent studies using single-cell transcriptomic analysis have reported several distinct clusters of neoplastic epithelial cells and cancer-associated fibroblasts in the pancreatic cancer tumor microenvironment. However, their molecular characteristics and biological significance have not been clearly elucidated due to intra- and inter-tumoral heterogeneity. METHODS We performed single-cell RNA sequencing using enriched non-immune cell populations from 17 pancreatic tumor tissues (16 pancreatic cancer and one high-grade dysplasia) and generated paired spatial transcriptomic data from seven patient samples. RESULTS We identified five distinct functional subclusters of pancreatic cancer cells and six distinct cancer-associated fibroblast subclusters. We deeply profiled their characteristics, and we found that these subclusters successfully deconvoluted most of the features suggested in bulk transcriptome analysis of pancreatic cancer. Among those subclusters, we identified a novel cancer cell subcluster, Ep_VGLL1, showing intermediate characteristics between the extremities of basal-like and classical dichotomy, despite its prognostic value. Molecular features of Ep_VGLL1 suggest its transitional properties between basal-like and classical subtypes, which is supported by spatial transcriptomic data. CONCLUSIONS This integrative analysis not only provides a comprehensive landscape of pancreatic cancer and fibroblast population, but also suggests a novel insight to the dynamic states of pancreatic cancer cells and unveils potential therapeutic targets.
Collapse
Affiliation(s)
- Seongryong Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Galam Leem
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Junjeong Choi
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yongjun Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Suho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Sang-Hee Nam
- Department of Internal Medicine, Graduate School of Yonsei University, Seoul, Republic of Korea
| | - Jin Su Kim
- Department of Internal Medicine, Graduate School of Yonsei University, Seoul, Republic of Korea
| | - Chan Hee Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Ho Kyoung Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei Cancer Center, Yonsei University College of Medicine, Pancreatobiliary Cancer Center, Severance Hospital, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Republic of Korea
| | - Kyoung Il Min
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Chang Moo Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei Cancer Center, Yonsei University College of Medicine, Pancreatobiliary Cancer Center, Severance Hospital, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, Republic of Korea.
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
558
|
Sini MC, Doro MG, Frogheri L, Zinellu A, Paliogiannis P, Porcu A, Scognamillo F, Delogu D, Santeufemia DA, Persico I, Palomba G, Maestrale GB, Cossu A, Palmieri G. Combination of mutations in genes controlling DNA repair and high mutational load plays a prognostic role in pancreatic ductal adenocarcinoma (PDAC): a retrospective real-life study in Sardinian population. J Transl Med 2024; 22:108. [PMID: 38280995 PMCID: PMC10821545 DOI: 10.1186/s12967-024-04923-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND Patients with pancreatic ductal adenocarcinoma (PDCA) carrying impaired mismatch repair mechanisms seem to have an outcome advantage under treatment with conventional chemotherapy, whereas the role for the tumor mutation burden on prognosis is controversial. In this study, we evaluated the prognostic role of the mutated genes involved in genome damage repair in a real-life series of PDAC patients in a hospital-based manner from the main Institution deputed to surgically treat such a disease in North Sardinia. METHODS A cohort of fifty-five consecutive PDAC patients with potentially resectable/border line resectable PDAC (stage IIB-III) or oligometastatic disease (stage IV) and tumor tissue availability underwent next-generation sequencing (NGS)-based analysis using a panel containing driver oncogenes and tumor suppressor genes as well as genes controlling DNA repair mechanisms. RESULTS Genes involved in the both genome damage repair (DR) and DNA mismatch repair (MMR) were found mutated in 17 (31%) and 15 (27%) cases, respectively. One fourth of PDAC cases (14/55; 25.5%) carried tumors presenting a combination of mutations in repair genes (DR and MMR) and the highest mutation load rates (MLR-H). After correction for confounders (surgery, adjuvant therapy, stage T, and metastasis), multivariate Cox regression analysis indicated that mutations in DR genes (HR = 3.0126, 95% CI 1.0707 to 8.4764, p = 0.0367) and the MLR (HR = 1.0018, 95%CI 1.0005 to 1.0032, p = 0.009) were significantly related to worse survival. CONCLUSIONS The combination of mutated repair genes and MLR-H, which is associated with a worse survival in our series of PDAC patients treated with conventional chemotherapy protocols, might become a predictive biomarker of response to immunotherapy in addition to its prognostic role in predicting survival.
Collapse
Affiliation(s)
- Maria Cristina Sini
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Maria Grazia Doro
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Laura Frogheri
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Panagiotis Paliogiannis
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Traversa La Crucca 3, 07100, Sassari, Italy
| | - Alberto Porcu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Traversa La Crucca 3, 07100, Sassari, Italy
| | - Fabrizio Scognamillo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Traversa La Crucca 3, 07100, Sassari, Italy
| | - Daniele Delogu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Traversa La Crucca 3, 07100, Sassari, Italy
| | | | - Ivana Persico
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Grazia Palomba
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Giovanni Battista Maestrale
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Antonio Cossu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Traversa La Crucca 3, 07100, Sassari, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Genetic Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy.
- Immuno-Oncology & Targeted Cancer Biotherapies, University of Sassari, Sassari, Italy.
| |
Collapse
|
559
|
Motobayashi H, Kitahata Y, Okada KI, Miyazawa M, Ueno M, Hayami S, Miyamoto A, Shimizu A, Sato M, Yoshimura T, Nakamura Y, Takemoto N, Nakai T, Hyo T, Matsumoto K, Yamaue H, Kawai M. Short-term serial circulating tumor DNA assessment predicts therapeutic efficacy for patients with advanced pancreatic cancer. J Cancer Res Clin Oncol 2024; 150:35. [PMID: 38277079 PMCID: PMC10817839 DOI: 10.1007/s00432-023-05594-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024]
Abstract
PURPOSE We investigated the potential clinical utility of short-term serial KRAS-mutated circulating cell-free tumor DNA (ctDNA) assessment for predicting therapeutic response in patients undergoing first-line chemotherapy for advanced pancreatic cancer. METHODS We collected 144 blood samples from 18 patients with locally advanced or metastatic cancer that were undergoing initial first-line chemotherapy of gemcitabine plus nab-paclitaxel (GEM plus nab-PTX). Analysis of KRAS-mutated ctDNA was quantified by digital droplet polymerase chain reaction (ddPCR) as mutant allele frequency (MAF). This study investigated pretreatment KRAS-mutated ctDNA status and ctDNA kinetics every few days (days 1, 3, 5 and 7) after initiation of chemotherapy and their potential as predictive indicators. RESULTS Of the 18 enrolled patients, an increase in KRAS-mutated ctDNA MAF values from day 0-7 after initiation of chemotherapy was significantly associated with disease progression (P < 0.001). Meanwhile, positive pretreatment ctDNA status (MAF ≥ 0.02%) (P = 0.585) and carbohydrate antigen 19-9 (CA19-9) values above the median (P = 0.266) were not associated with disease progression. In univariate analysis, this short-term increase in ctDNA MAF values (day 0-7) was found to be associated with significantly shorter progression free survival (PFS) (hazard ration [HR], 24.234; range, (2.761-212.686); P = 0.0002). CONCLUSION This short-term ctDNA kinetics assessment may provide predictive information to reflect real-time therapeutic response and lead to effective refinement of regimen in patients with advanced pancreatic cancer undergoing systemic chemotherapy.
Collapse
Affiliation(s)
- Hideki Motobayashi
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Yuji Kitahata
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan.
| | - Ken-Ichi Okada
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Motoki Miyazawa
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Masaki Ueno
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Shinya Hayami
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Atsushi Miyamoto
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Atsushi Shimizu
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Masatoshi Sato
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Tomohiro Yoshimura
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Yuki Nakamura
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Norio Takemoto
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Tomoki Nakai
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Takahiko Hyo
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Kyohei Matsumoto
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| | - Hiroki Yamaue
- Department of Cancer Immunology, Wakayama Medical University, Wakayama, Japan
| | - Manabu Kawai
- Second Department of Surgery, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8510, Japan
| |
Collapse
|
560
|
Bashiardes S, Christodoulou C. Orally Administered Drugs and Their Complicated Relationship with Our Gastrointestinal Tract. Microorganisms 2024; 12:242. [PMID: 38399646 PMCID: PMC10893523 DOI: 10.3390/microorganisms12020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Orally administered compounds represent the great majority of all pharmaceutical compounds produced for human use and are the most popular among patients since they are practical and easy to self-administer. Following ingestion, orally administered drugs begin a "perilous" journey down the gastrointestinal tract and their bioavailability is modulated by numerous factors. The gastrointestinal (GI) tract anatomy can modulate drug bioavailability and accounts for interpatient drug response heterogeneity. Furthermore, host genetics is a contributor to drug bioavailability modulation. Importantly, a component of the GI tract that has been gaining notoriety with regard to drug treatment interactions is the gut microbiota, which shares a two-way interaction with pharmaceutical compounds in that they can be influenced by and are able to influence administered drugs. Overall, orally administered drugs are a patient-friendly treatment option. However, during their journey down the GI tract, there are numerous host factors that can modulate drug bioavailability in a patient-specific manner.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Molecular Virology Department, Cyprus Institute of Neurology and Genetics, Iroon Avenue 6, Nicosia 2371, Cyprus;
| | | |
Collapse
|
561
|
Morizane C, Ueno M, Ikeda M, Sudo K, Hirashima Y, Kuroda M, Ueno S, Okusaka T, Furuse J. A Phase 2 study of nivolumab in combination with modified FOLFIRINOX for metastatic pancreatic cancer. BJC REPORTS 2024; 2:3. [PMID: 39516689 PMCID: PMC11524122 DOI: 10.1038/s44276-023-00028-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 11/16/2024]
Abstract
BACKGROUND Nivolumab with modified FOLFIRINOX (mFOLFIRINOX) may have additive antitumour effects while minimising chemotherapy cytotoxicity. We assessed the efficacy and safety of nivolumab+mFOLFIRINOX in metastatic pancreatic cancer. METHODS Thirty-one treatment-naïve patients aged ≥20 years with metastatic unresectable/recurrent pancreatic cancer (≥1 measurable lesion per Response Evaluation Criteria in Solid Tumours version 1.1) and Eastern Cooperative Oncology Group 0/1 score and life expectancy ≥90 days received nivolumab (480 mg, every 4 weeks) plus mFOLFIRINOX. The primary endpoint was objective response rate (ORR). Secondary endpoints included overall survival (OS), progression-free survival (PFS) and safety. RESULTS At the median follow-up of 13.4 months, the ORR was 32.3% (complete response 0%; partial response 32.3%) and the median duration of response was 7.4 (range: 3.5-21.9) months; the primary endpoint was not met. Median OS and PFS were 13.4 (95% confidence interval [CI]: 10.6-16.6) months and 7.4 (95% CI: 3.9-9.2) months, respectively. The 1-year survival rate was 54.8% (95% CI: 36.0%-70.3%). Drug-related serious adverse events were reported in 29.0% of the patients; 3.2% drug-related adverse events led to discontinuation, and none led to death within 30-day safety window. CONCLUSION Nivolumab+mFOLFIRINOX was tolerable in patients with metastatic pancreatic cancer. ORR and survival were comparable to previously reported data. (JapicCTI-184230).
Collapse
Affiliation(s)
- Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yoshinori Hirashima
- Clinical Development Headquarters, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Masataka Kuroda
- Clinical Development Headquarters, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Shinji Ueno
- Clinical Development Headquarters, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Mitaka, Japan
- Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
562
|
Mendes I, Vale N. Overcoming Microbiome-Acquired Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Biomedicines 2024; 12:227. [PMID: 38275398 PMCID: PMC10813061 DOI: 10.3390/biomedicines12010227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Gastrointestinal cancers (GICs) are one of the most recurrent diseases in the world. Among all GICs, pancreatic cancer (PC) is one of the deadliest and continues to disrupt people's lives worldwide. The most frequent pancreatic cancer type is pancreatic ductal adenocarcinoma (PDAC), representing 90 to 95% of all pancreatic malignancies. PC is one of the cancers with the worst prognoses due to its non-specific symptoms that lead to a late diagnosis, but also due to the high resistance it develops to anticancer drugs. Gemcitabine is a standard treatment option for PDAC, however, resistance to this anticancer drug develops very fast. The microbiome was recently classified as a cancer hallmark and has emerged in several studies detailing how it promotes drug resistance. However, this area of study still has seen very little development, and more answers will help in developing personalized medicine. PC is one of the cancers with the highest mortality rates; therefore, it is crucial to explore how the microbiome may mold the response to reference drugs used in PDAC, such as gemcitabine. In this article, we provide a review of what has already been investigated regarding the impact that the microbiome has on the development of PDAC in terms of its effect on the gemcitabine pathway, which may influence the response to gemcitabine. Therapeutic advances in this type of GIC could bring innovative solutions and more effective therapeutic strategies for other types of GIC, such as colorectal cancer (CRC), due to its close relation with the microbiome.
Collapse
Affiliation(s)
- Inês Mendes
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- School of Life and Environmental Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Edifício de Geociências, 5000-801 Vila Real, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
563
|
Patel H, Zanos T, Hewitt DB. Deep Learning Applications in Pancreatic Cancer. Cancers (Basel) 2024; 16:436. [PMID: 38275877 PMCID: PMC10814475 DOI: 10.3390/cancers16020436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Pancreatic cancer is one of the most lethal gastrointestinal malignancies. Despite advances in cross-sectional imaging, chemotherapy, radiation therapy, and surgical techniques, the 5-year overall survival is only 12%. With the advent and rapid adoption of AI across all industries, we present a review of applications of DL in the care of patients diagnosed with PC. A review of different DL techniques with applications across diagnosis, management, and monitoring is presented across the different pathological subtypes of pancreatic cancer. This systematic review highlights AI as an emerging technology in the care of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Hardik Patel
- Northwell Health—The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA;
| | - Theodoros Zanos
- Northwell Health—The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA;
| | - D. Brock Hewitt
- Department of Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA;
| |
Collapse
|
564
|
Hernández-Blanquisett A, Quintero-Carreño V, Martínez-Ávila MC, Porto M, Manzur-Barbur MC, Buendía E. Metastatic Pancreatic Cancer: Where Are We? Oncol Rev 2024; 17:11364. [PMID: 38304752 PMCID: PMC10830814 DOI: 10.3389/or.2023.11364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/20/2023] [Indexed: 02/03/2024] Open
Abstract
Pancreatic cancer is one of the most lethal neoplasms worldwide; it is aggressive in nature and has a poor prognosis. The overall survival rate for pancreatic cancer is low. Most patients present non-specific symptoms in the advanced stages, which generally leads to late diagnosis, at which point there is no option for curative surgery. The treatment of metastatic pancreatic cancer includes systemic therapy, in some cases radiotherapy, and more recently, molecular targeted therapies, which can positively impact cancer control and improve quality of life. This review provides an overview of the molecular landscape of pancreatic cancer based on the most recent literature, as well as current treatment options for patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Abraham Hernández-Blanquisett
- Cancer Institute, Hospital Serena del Mar, Cartagena, Colombia
- Clinical Oncology, Hospital Serena del Mar, Cartagena, Colombia
| | - Valeria Quintero-Carreño
- Cancer Institute, Hospital Serena del Mar, Cartagena, Colombia
- Pain and Palliative Care Department, Hospital Serena del Mar, Cartagena, Colombia
| | | | - María Porto
- Cancer Institute, Hospital Serena del Mar, Cartagena, Colombia
| | - María Carolina Manzur-Barbur
- Cancer Institute, Hospital Serena del Mar, Cartagena, Colombia
- Internal Medicine Department, Hospital Serena del Mar, Cartagena, Colombia
| | - Emiro Buendía
- Cancer Institute, Hospital Serena del Mar, Cartagena, Colombia
- Internal Medicine Department, Hospital Serena del Mar, Cartagena, Colombia
| |
Collapse
|
565
|
Ganju V, Marx G, Pattison S, Amaro-Mugridge NB, Zhao JT, Williams BRG, MacDiarmid JA, Brahmbhatt H. Phase I/IIa Trial in Advanced Pancreatic Ductal Adenocarcinoma Treated with Cytotoxic Drug-Packaged, EGFR-Targeted Nanocells and Glycolipid-Packaged Nanocells. Clin Cancer Res 2024; 30:304-314. [PMID: 37976042 DOI: 10.1158/1078-0432.ccr-23-1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/21/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE We assessed the safety and efficacy of an EGFR-targeted, super-cytotoxic drug, PNU-159682-packaged nanocells with α-galactosyl ceramide-packaged nanocells (E-EDV-D682/GC) in patients with advanced pancreatic ductal adenocarcinoma (PDAC) who had exhausted all treatment options. PATIENTS AND METHODS ENG9 was a first-in-man, single-arm, open-label, phase I/IIa, dose-escalation clinical trial. Eligible patients had advanced PDAC, Eastern Cooperative Oncology Group status 0 to 1, and failed all treatments. Primary endpoints were safety and overall survival (OS). RESULTS Of 25 enrolled patients, seven were withdrawn due to rapidly progressive disease and one patient withdrew consent. All 25 patients were assessed for toxicity, 24 patients were assessed for OS, which was also assessed for 17 patients completing one treatment cycle [evaluable subset (ES)]. Nineteen patients (76.0%) experienced at least one treatment-related adverse event (graded 1 to 2) resolving within hours. There were no safety concerns, dose reductions, patient withdrawal, or treatment-related deaths. Median OS (mOS) was 4.4 months; however, mOS of the 17 ES patients was 6.9 months [208 days; range, 83-591 days; 95.0% confidence interval (CI), 5.6-10.3 months] and mOS of seven patients who did not complete one cycle was 1.8 months (54 days; range, 21-72; 95.0% CI, 1.2-2.2 months). Of the ES, 47.1% achieved stable disease and one partial response. Ten subjects in the ES survived over 6 months, the longest 19.7 months. During treatments, 82.0% of the ES maintained stable weight. CONCLUSIONS E-EDV-D682/GC provided significant OS, minimal side effects, and weight stabilization in patients with advanced PDAC. Advanced PDAC can be safely treated with super-cytotoxic drugs via EnGeneIC Dream Vectors to overcome multidrug resistance.
Collapse
Affiliation(s)
- Vinod Ganju
- Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University Faculty of Medicine, Nursing and Health Sciences, Clayton, Victoria, Australia
- Peninsula and Southeast Oncology (PASO), Frankston Private Hospital, Frankston, Australia
| | - Gavin Marx
- Sydney Adventist Hospital, Sydney, New South Wales, Australia
| | | | | | | | - Bryan R G Williams
- Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University Faculty of Medicine, Nursing and Health Sciences, Clayton, Victoria, Australia
| | | | | |
Collapse
|
566
|
Tripathi S, Tabari A, Mansur A, Dabbara H, Bridge CP, Daye D. From Machine Learning to Patient Outcomes: A Comprehensive Review of AI in Pancreatic Cancer. Diagnostics (Basel) 2024; 14:174. [PMID: 38248051 PMCID: PMC10814554 DOI: 10.3390/diagnostics14020174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Pancreatic cancer is a highly aggressive and difficult-to-detect cancer with a poor prognosis. Late diagnosis is common due to a lack of early symptoms, specific markers, and the challenging location of the pancreas. Imaging technologies have improved diagnosis, but there is still room for improvement in standardizing guidelines. Biopsies and histopathological analysis are challenging due to tumor heterogeneity. Artificial Intelligence (AI) revolutionizes healthcare by improving diagnosis, treatment, and patient care. AI algorithms can analyze medical images with precision, aiding in early disease detection. AI also plays a role in personalized medicine by analyzing patient data to tailor treatment plans. It streamlines administrative tasks, such as medical coding and documentation, and provides patient assistance through AI chatbots. However, challenges include data privacy, security, and ethical considerations. This review article focuses on the potential of AI in transforming pancreatic cancer care, offering improved diagnostics, personalized treatments, and operational efficiency, leading to better patient outcomes.
Collapse
Affiliation(s)
- Satvik Tripathi
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Arian Mansur
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Harika Dabbara
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Christopher P. Bridge
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
567
|
Fan M, Deng G, Ma Y, Si H, Wang Z, Dai G. Survival outcome of different treatment sequences in patients with locally advanced and metastatic pancreatic cancer. BMC Cancer 2024; 24:67. [PMID: 38216928 PMCID: PMC10785544 DOI: 10.1186/s12885-024-11823-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Despite some therapeutic advances, improvement in survival rates of unresectable and/or metastatic pancreatic ductal adenocarcinoma (PDAC) has been minimal over recent decade. We aimed to evaluate the impact of different treatment sequences on clinical outcomes of advanced PDAC at our academic institution. METHODS In this single institution retrospective analysis, we assessed characteristics and survival rates of unresectable and/or metastatic pancreatic PDAC patients who started a systemic treatment between 01/2015 and 12/2021. Survival analyses were performed by Kaplan-Meier and Cox proportional hazards model. RESULTS The number of 285 patients received at least two lines of treatment, but only 137 patients were suitable for third-line treatment. Subgroup analysis showed that thirty-seven patients received A line (gemcitabine/nab-paclitaxel or nab-paclitaxel combined therapy to FOLFIRINOX) therapy, 37 patients received B line (nab-paclitaxel combined therapy to gemcitabine combined therapy to FOLFIRINOX) therapy, 21 patients received C line (nab-paclitaxel combined therapy to gemcitabine combined therapy to oxaliplatin or irinotecan combined therapy) therapy. Survival rates for different treatment lines were significantly different and median overall survival (OS) was 14.00, 18.00, and 14.00 months, respectively (p<0.05). CONCLUSION Our study provides real-world evidence for the effectiveness of different treatment sequences and underscores the treatment sequences on survival outcome when considering the entire management in advanced PDAC.
Collapse
Affiliation(s)
- Mengjiao Fan
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
- Department of oncology, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guochao Deng
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yue Ma
- Medical School of Chinese People's Liberation Army, Beijing, China
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haiyan Si
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhikuan Wang
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Guanghai Dai
- Department of oncology, The Fifth Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
568
|
Lumibao JC, Okhovat SR, Peck KL, Lin X, Lande K, Yomtoubian S, Ng I, Tiriac H, Lowy AM, Zou J, Engle DD. The effect of extracellular matrix on the precision medicine utility of pancreatic cancer patient-derived organoids. JCI Insight 2024; 9:e172419. [PMID: 38051586 PMCID: PMC10906458 DOI: 10.1172/jci.insight.172419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
The use of patient-derived organoids (PDOs) to characterize therapeutic sensitivity and resistance is a promising precision medicine approach, and its potential to inform clinical decisions is now being tested in several large multiinstitutional clinical trials. PDOs are cultivated in the extracellular matrix from basement membrane extracts (BMEs) that are most commonly acquired commercially. Each clinical site utilizes distinct BME lots and may be restricted due to the availability of commercial BME sources. However, the effect of different sources of BMEs on organoid drug response is unknown. Here, we tested the effect of BME source on proliferation, drug response, and gene expression in mouse and human pancreatic ductal adenocarcinoma (PDA) organoids. Both human and mouse organoids displayed increased proliferation in Matrigel compared with Cultrex and UltiMatrix. However, we observed no substantial effect on drug response when organoids were cultured in Matrigel, Cultrex, or UltiMatrix. We also did not observe major shifts in gene expression across the different BME sources, and PDOs maintained their classical or basal-like designation. Overall, we found that the BME source (Matrigel, Cultrex, UltiMatrix) does not shift PDO dose-response curves or drug testing results, indicating that PDO pharmacotyping is a robust approach for precision medicine.
Collapse
Affiliation(s)
- Jan C. Lumibao
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Shira R. Okhovat
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Kristina L. Peck
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Xiaoxue Lin
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Kathryn Lande
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Shira Yomtoubian
- Salk Institute for Biological Studies, La Jolla, California, USA
| | - Isabella Ng
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, and
| | - Hervé Tiriac
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, and
| | - Andrew M. Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, and
| | - Jingjing Zou
- Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health and Human Longevity Science, UCSD, San Diego, California, USA
| | | |
Collapse
|
569
|
Kubo T, Muramatsu J, Arihara Y, Murota A, Ishikawa K, Yoshida M, Nagashima H, Tamura F, Ikeda Y, Usami M, Ono M, Nakamura H, Watanabe D, Shibata T, Kasahara K, Sakurai A, Takada K. Clinical characterization of patients with gBRCA1/2 mutation-positive unresectable pancreatic cancer: a multicenter prospective study. Jpn J Clin Oncol 2024; 54:47-53. [PMID: 37791389 PMCID: PMC10773213 DOI: 10.1093/jjco/hyad131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Accumulating evidence has demonstrated platinum-based chemotherapy followed by maintenance therapy with a poly Adenosine diphosphate (ADP)-ribose polymerase inhibitor (olaparib) show benefits in unresectable pancreatic cancer with a germline (g)BRCA1/2 mutation. Evaluation of the germline BRCA1 and BRCA2 mutation is essential for making decisions on a treatment strategy for patients with unresectable pancreatic cancer. However, the detection rates of germline BRCA1 and BRCA2 mutations and efficacy of maintenance with olaparib remain undetermined, prospectively, in Japan. METHODS & RESULTS In this prospective analysis, the rate of germline BRCA1 and BRCA2 mutations and efficacy of chemotherapy were analyzed in 136 patients with pancreatic cancer who underwent BRACAnalysis® (85 patients) or FoundationOne® CDx (51 patients) between January 2020 and July 2022. A total of six patients (4.4%) had a germline BRCA1 and BRCA2 mutation. Five patients were treated with modified FOLFIRINOX and one with fluorouracil and oxaliplatin. All patients continued platinum-based chemotherapy for ˃4 months and were subsequently treated with olaparib as a maintenance therapy. The response rate to platinum-based chemotherapy in the germline BRCA1 and BRCA2 mutation-positive group was significantly better than that of the germline BRCA1 and BRCA2 mutation-negative group (66% vs 23%, P = 0.04). All patients harbouring a germline BRCA1 and BRCA2 mutation were able to switch to olaparib. The median progression-free survival using olaparib was 5.7 months (range 3.0-9.2). CONCLUSIONS The rate of germline BRCA1 and BRCA2 mutations found in patients with unresectable pancreatic cancer was comparable to those of previous studies.An analysis of germline BRCA1 and BRCA2 mutations has benefits for all patients with unresectable pancreatic cancer with regard to decisions on therapeutic strategies in a clinical practice setting.
Collapse
Affiliation(s)
- Tomohiro Kubo
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Joji Muramatsu
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yohei Arihara
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ayako Murota
- Department of Medical Genetics and Genomics, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuma Ishikawa
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Yoshida
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Fumito Tamura
- Department of Gastroenterology, Hokkaido Cancer Center, Sapporo, Japan
| | - Yuki Ikeda
- Department of Gastroenterology, Oji General Hospital, Tomakomai, Japan
| | - Makoto Usami
- Department of Medical Oncology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Michihiro Ono
- Department of Gastroenterology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Hajime Nakamura
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Gastroenterology, Otaru Ekisaikai Hospital, Otaru, Japan
| | - Daichi Watanabe
- Department of Gastroenterology, Japanese Red Cross Date Hospital, Date, Japan
| | - Takanori Shibata
- Department of Gastroenterology, Rumoi City Hospital, Rumoi, Japan
| | - Kaoru Kasahara
- Department of Gastroenterology, Hakodate Goryoukaku Hospital, Hakodate, Japan
| | - Akihiro Sakurai
- Department of Medical Genetics and Genomics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
570
|
Luo W, Wen T, Qu X. Tumor immune microenvironment-based therapies in pancreatic ductal adenocarcinoma: time to update the concept. J Exp Clin Cancer Res 2024; 43:8. [PMID: 38167055 PMCID: PMC10759657 DOI: 10.1186/s13046-023-02935-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumors. The tumor immune microenvironment (TIME) formed by interactions among cancer cells, immune cells, cancer-associated fibroblasts (CAF), and extracellular matrix (ECM) components drives PDAC in a more immunosuppressive direction: this is a major cause of therapy resistance and poor prognosis. In recent years, research has advanced our understanding of the signaling mechanism by which TIME components interact with the tumor and the evolution of immunophenotyping. Through revolutionary technologies such as single-cell sequencing, we have gone from simply classifying PDACs as "cold" and "hot" to a more comprehensive approach of immunophenotyping that considers all the cells and matrix components. This is key to improving the clinical efficacy of PDAC treatments. In this review, we elaborate on various TIME components in PDAC, the signaling mechanisms underlying their interactions, and the latest research into PDAC immunophenotyping. A deep understanding of these network interactions will contribute to the effective combination of TIME-based therapeutic approaches, such as immune checkpoint inhibitors (ICI), adoptive cell therapy, therapies targeting myeloid cells, CAF reprogramming, and stromal normalization. By selecting the appropriate integrated therapies based on precise immunophenotyping, significant advances in the future treatment of PDAC are possible.
Collapse
Affiliation(s)
- Wenyu Luo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
- Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
571
|
Nichetti F, Rota S, Ambrosini P, Pircher C, Gusmaroli E, Droz Dit Busset M, Pusceddu S, Sposito C, Coppa J, Morano F, Pietrantonio F, Di Bartolomeo M, Mariani L, Mazzaferro V, de Braud F, Niger M. NALIRIFOX, FOLFIRINOX, and Gemcitabine With Nab-Paclitaxel as First-Line Chemotherapy for Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. JAMA Netw Open 2024; 7:e2350756. [PMID: 38190183 PMCID: PMC10774994 DOI: 10.1001/jamanetworkopen.2023.50756] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/20/2023] [Indexed: 01/09/2024] Open
Abstract
Importance The NAPOLI 3 trial showed the superiority of fluorouracil, leucovorin, liposomal irinotecan, and oxaliplatin (NALIRIFOX) over the combination of gemcitabine and nab-paclitaxel (GEM-NABP) as first-line treatment of metastatic pancreatic ductal adenocarcinoma (PDAC). Analyses comparing NALIRIFOX and GEM-NABP with fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) have not yet been reported. Objective To derive survival, response, and toxic effects data from phase 3 clinical trials and compare NALIRIFOX, FOLFIRINOX, and GEM-NABP. Data Sources After a systematic search of PubMed, Scopus, Embase, and American Society of Clinical Oncology and European Society for Medical Oncology meetings' libraries, Kaplan-Meier curves were extracted from phase 3 clinical trials conducted from January 1, 2011, until September 12, 2023. Study Selection Phase 3 clinical trials that tested NALIRIFOX, FOLFIRINOX, or GEM-NABP as first-line treatment of metastatic PDAC and reported overall survival (OS) and progression-free survival (PFS) curves were selected. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses of Individual Participant Data reporting guidelines. Data Extraction And Synthesis Individual patient OS and PFS data were extracted from Kaplan-Meier plots of original trials via a graphic reconstructive algorithm. Overall response rates (ORRs) and grade 3 or higher toxic effects rates were also collected. A pooled analysis was conducted, and results were validated via a network meta-analysis. Main Outcomes and Measures The primary end point was OS. Secondary outcomes included PFS, ORR, and toxic effects rates. Results A total of 7 trials with data on 2581 patients were analyzed, including 383 patients treated with NALIRIFOX, 433 patients treated with FOLFIRINOX, and 1756 patients treated with GEM-NABP. Median PFS was longer in patients treated with NALIRIFOX (7.4 [95% CI, 6.1-7.7] months) or FOLFIRINOX (7.3 [95% CI, 6.5-7.9] months; [HR], 1.21 [95% CI, 0.86-1.70]; P = .28) compared with patients treated with GEM-NABP (5.7 [95% CI, 5.6-6.1] months; HR vs NALIRIFOX, 1.45 [95% CI, 1.22-1.73]; P < .001). Similarly, GEM-NABP was associated with poorer OS (10.4 [95% CI, 9.8-10.8]; months) compared with NALIRIFOX (HR, 1.18 [95% CI, 1.00-1.39]; P = .05], while no difference was observed between FOLFIRINOX (11.7 [95% CI, 10.4-13.0] months) and NALIRIFOX (11.1 [95% CI, 10.1-12.3] months; HR, 1.06 [95% CI, 0.81-1.39]; P = .65). There were no statistically significant differences in ORR among NALIRIFOX (41.8%), FOLFIRINOX (31.6%), and GEM-NABP (35.0%). NALIRIFOX was associated with lower incidence of grade 3 or higher hematological toxic effects (eg, platelet count decreased 1.6% vs 11.8% with FOLFIRINOX and 10.8% with GEM-NABP), but higher rates of severe diarrhea compared with GEM-NABP (20.3% vs 15.7%). Conclusions and Relevance In this systematic review and meta-analysis, NALIRIFOX and FOLFIRINOX were associated with similar PFS and OS as first-line treatment of advanced PDAC, although NALIRIFOX was associated with a different toxicity profile. Careful patient selection, financial toxic effects consideration, and direct comparison between FOLFIRINOX and NALIRIFOX are warranted.
Collapse
Affiliation(s)
- Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases and German Cancer Research Center , Heidelberg, Germany
| | - Simone Rota
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Pircher
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eleonora Gusmaroli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Droz Dit Busset
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Sposito
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Jorgelina Coppa
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luigi Mariani
- Department of Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Vincenzo Mazzaferro
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
572
|
Kong Q, Teng F, Li H, Chen Z. Radical resection benefits patients suffering pancreatic ductal adenocarcinoma with liver oligometastases. Ann Surg Treat Res 2024; 106:51-60. [PMID: 38205094 PMCID: PMC10774701 DOI: 10.4174/astr.2024.106.1.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 01/12/2024] Open
Abstract
Purpose Whether patients suffering liver oligometastases from pancreatic ductal adenocarcinoma (LOPDA) should undergo surgical treatment remains controversial. Methods PubMed and Embase databases were systematically reviewed until 2023 June. Survival data were collected from the Kaplan-Meier curves. Safety and survival were evaluated using primary outcomes such as 1-year, 3-year, and 5-year survival rates, and 30-day mortality and morbidity. A subgroup meta-analysis was conducted to compare survival rates post-synchronous resection and resection post-neoadjuvant chemotherapy in LOPDA. Results Our analysis of 15 studies involving 1,818 patients (surgical group, 648 and nonsurgical group, 1,170) indicates that radical hepatectomy for LOPDA notably improved 1-year (odds ratio [OR], 3.24; 95% confidence interval [CI], 2.45-4.28; P < 0.001), 3-year (OR, 5.74; 95% CI, 3.36-8.90; P < 0.001), and 5-year (OR, 4.89; 95% CI, 2.56-9.35; P < 0.001) overall survival (OS) rates. A separate analysis of 6 studies with 750 patients demonstrated the safety of LOPDA surgery, with no increase in postoperative complications (P = 0.26 for overall morbidity and P = 0.99 for mortality) compared to the patients with no metastatic disease from the pancreatic ductal adenocarcinoma (NMPDA) group. The NMPDA group showed superior 1-year and 3-year OS rates, but not 5-year OS rates compared to the LOPDA group. Conclusion Surgical treatment apparently offers a survival advantage to LOPDA by comparing with nonsurgical groups in 1-, 3-, and 5-year OS rates. Radical resection for LOPDA is a safe treatment without more postoperative complications than NMPDA.
Collapse
Affiliation(s)
- Qingyan Kong
- Division of Hepatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Teng
- Division of Hepatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Li
- Division of Hepatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zheyu Chen
- Division of Hepatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
573
|
Cruz MS, Tintelnot J, Gagliani N. Roles of microbiota in pancreatic cancer development and treatment. Gut Microbes 2024; 16:2320280. [PMID: 38411395 PMCID: PMC10900280 DOI: 10.1080/19490976.2024.2320280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.
Collapse
Affiliation(s)
- Mariana Santos Cruz
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Joseph Tintelnot
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
574
|
Wang LT, Yang J, Wu J, Lu WJ. Combined Therapy of Chemotherapy and Radiofrequency Ablation for Pancreatic Cancer Patients With Metachronous Hepatic Metastatic Lesions After Radical Pancreatic Resection. Cancer Control 2024; 31:10732748241274559. [PMID: 39150275 PMCID: PMC11329956 DOI: 10.1177/10732748241274559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
PURPOSE Hepatic metastasis frequently occurs in patients who have undergone radical pancreatic resection for pancreatic cancer. Besides chemotherapy, various local treatment approaches targeting hepatic lesions have been explored. However, research on radiofrequency ablation (RFA) as a localized therapy for hepatic metastasis is limited. Therefore, we conducted this retrospective study to provide clinical evidence. METHODS This is a single-center, retrospective, cohort study. After radical pancreaticoduodenectomy, 32 patients developed metachronous hepatic metastasis with fewer than 3 lesions, the largest of which was less than 3 cm in diameter. These patients underwent combined treatment with chemotherapy and RFA. After 8 weeks of chemotherapy, patients received RFA for hepatic lesions. Additional chemotherapy was administered, and the patients' tumor status and survival were monitored. The primary endpoint of this study was overall survival (OS). Factors affecting OS were analyzed using the Cox risk model. RESULTS Among the 32 patients, the mean OS was 28.4 months. Univariate and multivariate Cox regression analysis revealed that the time (in months) of liver metastasis (HR = 0.04, 95% CI: 0.01 to 0.19; P < 0.001), the number of liver metastases (HR = 7.08, 95% CI: 1.85 to 27.08, P = 0.004), and PD (progressive disease) response to the second round of chemotherapy (HR = 29.50, 95% CI: 1.46 to 597.27; P = 0.027) were independent predictors of poorer survival. CONCLUSION Combined therapy with RFA and chemotherapy is safe in patients with hepatic metastasis after radical pancreaticoduodenectomy. Early recurrence (≤12 months), three liver metastatic lesions, and a poor response to the second round of chemotherapy were associated with poor survival.
Collapse
Affiliation(s)
- L. T. Wang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - J. Yang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - J. Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - W. J. Lu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
575
|
Lundy J. A Humanized Patient-Derived Xenograft Model for Pancreatic Cancer. Methods Mol Biol 2024; 2806:91-100. [PMID: 38676798 DOI: 10.1007/978-1-0716-3858-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Pancreatic cancer is associated with a high mortality rate, and there are still very few effective treatment options. Patient-derived xenografts have proven to be invaluable preclinical disease models to study cancer biology and facilitate testing of novel therapeutics. However, the severely immune-deficient mice used to generate standard models lack any functional immune system, thereby limiting their utility as a tool to investigate the tumor-immune cell interface. This chapter will outline a method for establishment of "humanized" patient-derived xenografts, which are reconstituted with human immune cells to imitate the immune-rich microenvironment of pancreatic cancer.
Collapse
Affiliation(s)
- Joanne Lundy
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
- Department of Surgery, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
- Peninsula Clinical School, Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
576
|
Mody J, Kamgar M. Pancreatic Adenocarcinoma with Co-Occurrence of KRAS and EGFR Mutations: Case Report and Literature Review. Case Rep Oncol 2024; 17:399-406. [PMID: 38435447 PMCID: PMC10907001 DOI: 10.1159/000536552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Mutation in Kristin ras sarcoma virus (KRAS) oncogene is the main driver in pancreatic ductal adenocarcinoma (PDAC) and is present in nearly 90% of patients with PDAC. Epidermal growth factor receptor (EGFR) mutation is rare in PDAC and is mostly present in the absence of KRAS mutation. Co-occurrence of KRAS and EGFR mutations is extremely rare, and the value of EGFR inhibition in these cases is unknown. Case Presentation Here, we present a case of metastatic PDAC with co-occurrence of KRAS G12V and EGFR L730R. Despite primary resistance to folinic acid, fluorouracil, irinotecan, oxaliplatin, and gemcitabine/nab-paclitaxel, this patient had a biochemical response (decrease in carbohydrate antigen 19-9) and disease control of 7 months on gemcitabine/erlotinib (an EGFR inhibitor). This outcome is remarkable in the late-line PDAC treatment setting and is unusual after the progression of the tumor on gemcitabine/nab-paclitaxel chemotherapy. Conclusion This case suggests that gemcitabine/erlotinib could be an effective treatment in patients with PDAC and co-occurrence of EGFR and KRAS mutations.
Collapse
Affiliation(s)
- Juhi Mody
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mandana Kamgar
- Medical College of Wisconsin and The LaBahn Pancreatic Cancer Program, Milwaukee, WI, USA
| |
Collapse
|
577
|
Ariake K, Okada T, Tsuchiya H, Kuboki D, Maemura K, Okada Y, Ichikawa H, Tachibana T, Akazawa N, Abe T, Kakita T, Oikawa M, Tsuchiya T. Postoperative Carbohydrate Antigen 19-9 Level as a Good Indicator of Ineffective Response to the Currently Recommended S-1 Adjuvant Chemotherapy for Pancreatic Ductal Adenocarcinoma: A Single-Center, Retrospective Study. Ann Surg Oncol 2024; 31:525-534. [PMID: 37851194 DOI: 10.1245/s10434-023-14440-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
PURPOSE The intensity of adjuvant treatment for pancreatic ductal adenocarcinomas (PDACs) has not been stratified according to the risk after resection. This study was designed to identify patients with PDACs in whom the current S-1 adjuvant treatment is ineffective. METHODS This single-center, retrospective study included patients who underwent pancreatectomy for PDACs from 2009 to 2020 at Sendai Open Hospital and were receiving S-1 adjuvant treatment. The independent risk factors for recurrence and survival were determined by using a Cox proportional hazards regression model. The effects of S-1 adjuvant treatment and detailed patterns of recurrence were evaluated in patients with high-risk factors. RESULTS Overall, 118 patients with PDAC received S-1 adjuvant treatment. Postoperative nonnormalized carbohydrate antigen (CA19-9) was a predictive risk factor for recurrence (p < 0.010; hazard ratio [HR], 3.87; 95% confidence interval [CI], 2.26-6.62) and survival (p = 0.008; HR, 2.25; 95% CI, 1.24-4.11) after S-1 adjuvant treatment. In 24 patients with nonnormalized postoperative CA19-9, S-1 monotherapy was ineffective in preventing recurrence, even during the treatment period, compared with that noted in patients who did not receive adjuvant treatment. The recurrence rate during adjuvant treatment was 41.7%; in all cases, recurrence was caused by distant metastasis. The total recurrence rate was up to 95.8%, and distant recurrence was especially frequent. CONCLUSIONS The current S-1 adjuvant treatment regimen is ineffective for patients with postoperative nonnormalized CA19-9. The postoperative CA19-9 level may be a good indicator for further aggressive treatment. This study may lead to further discussions on intensity stratification of adjuvant treatments for PDAC.
Collapse
Affiliation(s)
- Kyohei Ariake
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan.
| | - Takaho Okada
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Haruyuki Tsuchiya
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Daiki Kuboki
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Kimiya Maemura
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Yuki Okada
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Hidetaka Ichikawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Tomoyoshi Tachibana
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Naoya Akazawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Tomoya Abe
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Tetsuya Kakita
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Masaya Oikawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| | - Takashi Tsuchiya
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai, Japan
| |
Collapse
|
578
|
Dabiri R, Rashid MU, Khan OS, Jehanzeb S, Alomari M, Zafar H, Zahid E, Rahman AU, Karam A, Ahmad S. Immune modulators for pancreatic ductal adenocarcinoma therapy. IMMUNE LANDSCAPE OF PANCREATIC CANCER DEVELOPMENT AND DRUG RESISTANCE 2024:103-129. [DOI: 10.1016/b978-0-443-23523-8.00021-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
579
|
van Eijck CWF, Strijk G, Vietsch EE, van der Sijde F, Verheij M, Mustafa DAM, Vink M, Aerts JGJV, van Eijck CHJ, Willemsen M. FOLFIRINOX chemotherapy modulates the peripheral immune landscape in pancreatic cancer: Implications for combination therapies and early response prediction. Eur J Cancer 2024; 196:113440. [PMID: 37988843 DOI: 10.1016/j.ejca.2023.113440] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND FOLFIRINOX chemotherapy has improved outcomes for pancreatic cancer patients, but poor long-term survival outcomes and high toxicity remain challenges. This study investigates the impact of FOLFIRINOX on plasma proteins and peripheral immune cells to guide immune-based combination therapies and, ideally, to identify a potential biomarker to predict early disease progression during FOLFIRINOX. METHODS Blood samples were collected from 86 pancreatic cancer patients before and two weeks after the first FOLFIRINOX cycle and subjected to comprehensive immune cell and proteome profiling. Principal Component Analysis and Linear Mixed Effect Regression models were used for data analysis. FOLFIRINOX efficacy was radiologically evaluated after the fourth cycle. RESULTS One cycle of FOLFIRINOX diminished tumour-cell-related pathways and enhanced pathways related to immune activation, illustrated by an increase in pro-inflammatory IL-18, IL-15, and TNFRSF4. Similarly, FOLFIRINOX promoted the activation of CD4 + and CD8 + T cells, the proliferation of NK(T), and the activation of antigen-presenting cells. Furthermore, high pre-treatment levels of VEGFA and PRDX3 and an elevation in FCRL3 levels after one cycle predicted early progression under FOLFIRINOX. Finally, patients with progressive disease exhibited high levels of inhibitory markers on B cells and CD8 + T cells, while responding patients exhibited high levels of activation markers on CD4 + and CD8 + T cell subsets. CONCLUSION FOLFIRINOX has immunomodulatory effects, providing a foundation for clinical trials exploring immune-based combination therapies that harness the immune system to treat pancreatic cancer. In addition, several plasma proteins hold potential as circulating predictive biomarkers for early prediction of FOLFIRINOX response in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Gaby Strijk
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Fleur van der Sijde
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Maaike Verheij
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Madelief Vink
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Joachim G J V Aerts
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marcella Willemsen
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Pulmonary Medicine, Erasmus University Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
580
|
Khalil MA, Jafri SAA, Zahra Naqvi ME, Jauhar I, Kumar V, Ali SMS, Khalil S, Raheem A, kumar R, Haseeb A, Shafique MA. Impact of Cachexia on Chemotherapy Efficacy and Survival in Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancer Control 2024; 31:10732748241292784. [PMID: 39397738 PMCID: PMC11483828 DOI: 10.1177/10732748241292784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Pancreatic cancer (PC) is a significant cause of cancer-related mortality, with limited curative options and high rates of cachexia, a debilitating syndrome associated with poor prognosis. While previous research has linked sarcopenia to poor outcomes in PC, the correlation between cachexia and treatment outcomes remains underexplored. This meta-analysis aims to investigate the association between cachexia and overall survival and time to treatment failure in advanced PC patients undergoing first-line chemotherapy. METHOD A systematic search of electronic databases was conducted following PRISMA guidelines. Eligible studies compared cachexic and non-cachexic PC patients, reporting outcomes of observed survival or time to treatment failure. Data extraction and analysis were performed using Comprehensive Meta-Analysis Version 3.3, employing random-effects models and sensitivity analyses to assess heterogeneity and bias. RESULTS Seven observational studies involving 2834 PC patients were included. The incidence of cachexia was 45% (95% CI: 0.27-0.65), with a higher prevalence in East Asian populations. Cachexic patients experienced significantly earlier treatment failure (SDM: -2.22, 95% CI: -2.6 to -1.7, P = 0.0001) and higher mortality risk (HR: 2.02, 95% CI: 1.17-3.48, P = 0.011) compared to non-cachexic patients. Overall survival was lower in cachexic patients (SDM: -2.34, 95% CI: -3.7 to -0.90, P = 0.001), with considerable heterogeneity across studies. Meta-regression analysis revealed significant differences between countries but insignificant correlations with age. CONCLUSION Cachexia is associated with reduced overall survival, early chemotherapy failure, and elevated mortality in advanced PC patients undergoing first-line chemotherapy. Recognition and management of cachexia are crucial for optimizing treatment outcomes and improving patient survival. Future research should focus on prospective studies to better understand the impact of cachexia on treatment response and develop tailored interventions to mitigate its adverse effects.
Collapse
Affiliation(s)
| | - Syed Ahsan Ali Jafri
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - Midhat E. Zahra Naqvi
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - Iman Jauhar
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - Vijay Kumar
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | | | - Sara Khalil
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - Abdul Raheem
- Department of Medicine, Baqai Medical University, Karachi, Pakistan
| | - Ritesh kumar
- Department of Medicine, Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - Abdul Haseeb
- Department of Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | | |
Collapse
|
581
|
Brown ZJ, Pawlik TM. Top Hepatobiliary Articles from 2022 to Inform Your Cancer Practice. Ann Surg Oncol 2024; 31:75-80. [PMID: 37843669 DOI: 10.1245/s10434-023-14420-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
Until recently, there have been only modest therapeutic advances in the treatment of hepatobiliary malignancies. However, the introduction of immune checkpoint inhibitors in combination with targeted therapy or chemotherapy has changed the therapeutic landscape of hepatocellular carcinoma and biliary tract cancers. As such, revisions have been made to guidelines reflecting therapeutic advances for patients who can be considered for surgical options including resection and liver transplantation. This article highlights recently published studies that have impacted both the oncological and surgical approach to the treatment of patients with hepatobiliary malignancies.
Collapse
Affiliation(s)
- Zachary J Brown
- Division of Surgical Oncology, Department of Surgery, NYU Langone Health, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, The Urban Meyer III and Shelley Meyer Chair for Cancer Research, The Ohio State University Wexner Medical Center, James Cancer Hospital, Columbus, OH, USA.
| |
Collapse
|
582
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a rising incidence and is one of the most lethal human malignancies. Much is known regarding the biology and pathophysiology of PDAC, but translating this knowledge to the clinic to improve patient outcomes has been challenging. In this Review, we discuss advances and practice-changing trials for PDAC. We briefly review therapeutic failures as well as ongoing research to refine the standard of care, including novel biomarkers and clinical trial designs. In addition, we highlight contemporary areas of research, including poly(ADP-ribose) polymerase inhibitors, KRAS-targeted therapies and immunotherapies. Finally, we discuss the future of pancreatic cancer research and areas for improvement in the next decade.
Collapse
Affiliation(s)
- Z Ian Hu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
583
|
Zironda A, Zhang C, Day C, McWilliams RR, Starlinger P, Warner SG, Smoot RL, Cleary SP, Kendrick ML, Truty MJ, Thiels CA. Early vs conventional onset pancreatic ductal adenocarcinoma: analysis of surgical and oncologic outcomes in patients undergoing curative intent resection. HPB (Oxford) 2024; 26:145-153. [PMID: 37752029 DOI: 10.1016/j.hpb.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) impacts patients in their 60s, but its incidence in younger patients is increasing. We hypothesize that younger patients may have worse oncologic outcomes. METHODS Patients who underwent curative pancreatic resection for PDAC between January 2011 and December 2021 at a single institution were analyzed. Early-onset pancreatic cancer (EOPC) was defined as pancreatic cancer diagnosed in patients ≤50 years. Clinical and survival outcomes were compared between EOPC and Conventional Onset Pancreas Cancer (COPC). RESULTS A total of 1133 patients were identified, 65 (5.7%) were EOPC. Preoperative patient characteristics including sex, smoking status, alcohol habitus, diabetes mellitus, CA 19-9, and neoadjuvant therapy were similar between EOPC and COPC (p > 0.05). EOPC patients were more likely non-white (p = 0.03), had lower ASA scores (p = 0.02) and larger median tumor size (33 vs 28 mm, p = 0.04), but had similar pathological stages and rate of R0 resections (p > 0.05). Postoperative outcomes were similar (p > 0.05). There was no statistically significant difference in overall (HR 0.93, CI 0.64, 1.33; p = 0.68) or recurrence free (HR 1.05, CI 0.75, 1.48; p = 0.77) survival between the EOPC and COPC after adjusting for significant factors. CONCLUSION Patients with EOPC who underwent surgical resection had similar oncological outcomes compared to patients with COPC.
Collapse
Affiliation(s)
- Andrea Zironda
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Chi Zhang
- Department of Surgery, Mayo Clinic, Phoenix, AZ, USA; Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Courtney Day
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | | | - Patrick Starlinger
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Susanne G Warner
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Rory L Smoot
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Sean P Cleary
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Micheal L Kendrick
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Mark J Truty
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA
| | - Cornelius A Thiels
- Hepatobiliary and Pancreas Surgery Division, Mayo Clinic, Rochester MN, USA.
| |
Collapse
|
584
|
Pourali G, Donyadideh G, Mehrabadi S, Hamid F, Hassanian SM, Ferns GA, Khazaei M, Avan A. Clinical practice guidelines for interventional treatment of pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:345-373. [DOI: 10.1016/b978-0-443-19142-8.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
585
|
Buckley CW, O’Reilly EM. Next-generation therapies for pancreatic cancer. Expert Rev Gastroenterol Hepatol 2024; 18:55-72. [PMID: 38415709 PMCID: PMC10960610 DOI: 10.1080/17474124.2024.2322648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
INTRODUCTION Pancreas ductal adenocarcinoma (PDAC) is a frequently lethal malignancy that poses unique therapeutic challenges. The current mainstay of therapy for metastatic PDAC (mPDAC) is cytotoxic chemotherapy. NALIRIFOX (liposomal irinotecan, fluorouracil, leucovorin, oxaliplatin) is an emerging standard of care in the metastatic setting. An evolving understanding of PDAC pathogenesis is driving a shift toward targeted therapy. Olaparib, a poly-ADP-ribose polymerase (PARP) inhibitor, has regulatory approval for maintenance therapy in BRCA-mutated mPDAC along with other targeted agents receiving disease-agnostic approvals including for PDAC with rare fusions and mismatch repair deficiency. Ongoing research continues to identify and evaluate an expanding array of targeted therapies for PDAC. AREAS COVERED This review provides a brief overview of standard therapies for PDAC and an emphasis on current and emerging targeted therapies. EXPERT OPINION There is notable potential for targeted therapies for KRAS-mutated PDAC with opportunity for meaningful benefit for a sizable portion of patients with this disease. Further, emerging approaches are focused on novel immune, tumor microenvironment, and synthetic lethality strategies.
Collapse
Affiliation(s)
- Conor W. Buckley
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Eileen M. O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Weill Cornell Medicine, New York, USA
| |
Collapse
|
586
|
Yun WG, Han Y, Lee M, Cho YJ, Jung HS, Kwon W, Jang JY. Efficacy of local treatment for isolated liver metastasis after pancreatectomy in pancreatic ductal adenocarcinoma: A propensity score-matched analysis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:50-60. [PMID: 37800313 DOI: 10.1002/jhbp.1352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
BACKGROUND Although attempts of local treatment for isolated liver recurrence in patients with pancreatic ductal adenocarcinoma (PDAC) have increased, the efficacy remains unclear. Therefore, we aimed to evaluate the effect of local treatment for recurrent liver lesions after pancreatectomy on the survival of patients with PDAC. METHODS Patients who were diagnosed with isolated liver recurrence after pancreatectomy at a high-volume center were included. We classified these patients based on the treatment options after recurrence and performed propensity score matching to minimize confounding. RESULTS Median with interquartile range survival after recurrence was significantly longer for patients who underwent local treatment for recurrent liver lesions plus chemotherapy (22.0 [17.0-29.0] months) than those treated with chemotherapy alone (13.0 [7.0-21.0] months, p = .027). In multivariate analysis, not only local treatment for recurrent liver lesions plus chemotherapy (hazard ratio [95% confidence interval], 0.55 [0.32-0.94]; p = .030) but also indicators for systemically controlled tumor such as late recurrence (0.57 [0.35-0.92]; p = .021), chemotherapy for ≥6 months (0.25 [0.15-0.42]; p < .001), and disease control by chemotherapy (0.36 [0.22-0.60]; p < .001) were identified as favorable prognostic factors. CONCLUSIONS PDAC patients with stable recurrent liver lesions should be considered a candidate for local treatment.
Collapse
Affiliation(s)
- Won-Gun Yun
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Mirang Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jae Cho
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
587
|
Kanda T, Wakiya T, Ishido K, Kimura N, Nagase H, Yoshida E, Nakagawa J, Matsuzaka M, Niioka T, Sasaki Y, Hakamada K. Noninvasive Computed Tomography-Based Deep Learning Model Predicts In Vitro Chemosensitivity Assay Results in Pancreatic Cancer. Pancreas 2024; 53:e55-e61. [PMID: 38019604 DOI: 10.1097/mpa.0000000000002270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
OBJECTIVES We aimed to predict in vitro chemosensitivity assay results from computed tomography (CT) images by applying deep learning (DL) to optimize chemotherapy for pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Preoperative enhanced abdominal CT images and the histoculture drug response assay (HDRA) results were collected from 33 PDAC patients undergoing surgery. Deep learning was performed using CT images of both the HDRA-positive and HDRA-negative groups. We trimmed small patches from the entire tumor area. We established various prediction labels for HDRA results with 5-fluorouracil (FU), gemcitabine (GEM), and paclitaxel (PTX). We built a predictive model using a residual convolutional neural network and used 3-fold cross-validation. RESULTS Of the 33 patients, effective response to FU, GEM, and PTX by HDRA was observed in 19 (57.6%), 11 (33.3%), and 23 (88.5%) patients, respectively. The average accuracy and the area under the receiver operating characteristic curve (AUC) of the model for predicting the effective response to FU were 93.4% and 0.979, respectively. In the prediction of GEM, the models demonstrated high accuracy (92.8%) and AUC (0.969). Likewise, the model for predicting response to PTX had a high performance (accuracy, 95.9%; AUC, 0.979). CONCLUSIONS Our CT patch-based DL model exhibited high predictive performance in projecting HDRA results. Our study suggests that the DL approach could possibly provide a noninvasive means for the optimization of chemotherapy.
Collapse
Affiliation(s)
- Taishu Kanda
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Taiichi Wakiya
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Keinosuke Ishido
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Norihisa Kimura
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Hayato Nagase
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | - Eri Yoshida
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| | | | | | | | - Yoshihiro Sasaki
- Medical Informatics, Hirosaki University Hospital, Hirosaki, Japan
| | - Kenichi Hakamada
- From the Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki City
| |
Collapse
|
588
|
Ben-Ammar I, Rousseau A, Nicolle R, Tarabay A, Boige V, Valery M, Pudlarz T, Malka D, Gelli M, Fernandez-De-Sevilla E, Fuerea A, Tanguy ML, Rouleau E, Barbe R, Mathieu JRR, Jaulin F, Smolenschi C, Hollebecque A, Ducreux M, Boileve A. Precision medicine for KRAS wild-type pancreatic adenocarcinomas. Eur J Cancer 2024; 197:113497. [PMID: 38134480 DOI: 10.1016/j.ejca.2023.113497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND KRAS mutation is the most common molecular alteration in pancreatic adenocarcinoma (PDAC), and around 10% of patients harbor KRAS wild-type tumors (KRASWT). METHODS A retrospective chart review of clinical/molecular data was performed including all PDAC patients with a determined KRAS status (tumor molecular profiling on tissue or liquid biopsy). RESULTS 342 patients were included with 54 KRASWT PDAC (16%) compared to 288 patients with KRASm PDAC. Median age was 61 years [IQR:54.0;67.0] and 164 pts (48%) were female. At diagnosis, KRASWT patients (63%) were more frequently diagnosed at a non-metastatic stage compared to KRASm patients (41%) (p = 0.003). Regarding metastatic sites, liver was less frequent in KRASWT (39%, p < 0.0001). Median overall survival (mOS) from initial diagnosis was significantly higher in the KRASWT group compared to KRASm (50.8 months, CI95% [32.0-NR] vs 21.1 months, CI95% [18.9-23.4] (p < 0.004 after adjustment on age, ECOG and stage at diagnosis). In first-line systemic treatment, (mostly FOLFIRINOX) progression-free survival (PFS) was also higher in KRASWT. Based on ESCAT classification, a putative actionable alteration (ESCAT I-III) was identified in 19 (36%) KRASWT pts and 46 (16%) KRASm patients (p < 0.0001) with more alterations in FGFR2, BRAF(V600E), NRTK and more MSI tumors. KRASWT harbored also fewer alterations in TP53, CDKN2A, and SMAD4. 12 KRASWT patients received a molecularly-matched treatment with clinical benefit and improved outcomes compared to KRASm patients. CONCLUSIONS KRASWT patients display distinct disease characteristics and outcomes with prolonged overall survival. KRASWT patients also harbor more actionable molecular alterations, leading to higher survival rates after receiving molecularly matched treatments.
Collapse
Affiliation(s)
- Imen Ben-Ammar
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; Sorbonne Université, Faculté de Médecine, 75005 Paris, France
| | - Adrien Rousseau
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; Oncostat INSERM U1018, Gustave Roussy, Ligue Contre le Cancer, Université Paris-Saclay, Villejuif, France; Gustave Roussy, Département de Biostatistiques et D'épidémiologie, Université Paris-Saclay, Villejuif, France
| | - Rémy Nicolle
- Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Université Paris Cité, Paris, France
| | - Anthony Tarabay
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France
| | - Valérie Boige
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France
| | - Marine Valery
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France
| | - Thomas Pudlarz
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France
| | - David Malka
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France
| | | | | | - Alina Fuerea
- Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Université Paris Cité, Paris, France
| | - Marie-Laure Tanguy
- Gustave Roussy, Département de Biostatistiques et D'épidémiologie, Université Paris-Saclay, Villejuif, France
| | - Etienne Rouleau
- Gustave Roussy, Département de Génétique Médicale, 94800 Villejuif, France
| | - Rémy Barbe
- Gustave Roussy, Département de Radiologie, 94800 Villejuif, France
| | | | - Fanny Jaulin
- INSERM U1279, Gustave Roussy, 94800 Villejuif, France; Université Paris Saclay, 91471 Orsay, France
| | - Cristina Smolenschi
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; Gustave Roussy, DITEP, 94800 Villejuif, France
| | - Antoine Hollebecque
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; Gustave Roussy, DITEP, 94800 Villejuif, France
| | - Michel Ducreux
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; INSERM U1279, Gustave Roussy, 94800 Villejuif, France; Université Paris Saclay, 91471 Orsay, France
| | - Alice Boileve
- Gustave Roussy, Département de Médecine, 94800 Villejuif, France; INSERM U1279, Gustave Roussy, 94800 Villejuif, France; Université Paris Saclay, 91471 Orsay, France.
| |
Collapse
|
589
|
de Ortiz de Choudens S, Visotcky A, Banerjee A, Aldakkak M, Tsai S, Evans DB, Christians KK, Clarke CN, George B, Shreenivas A, Kamgar M, Chakrabarti S, Dua KS, Khan AH, Madhavan S, Erickson BA, Hall WA. Characterization of an oligometastatic state in patients with metastatic pancreatic adenocarcinoma undergoing systemic chemotherapy. Cancer Med 2024; 13:e6582. [PMID: 38140796 PMCID: PMC10807686 DOI: 10.1002/cam4.6582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/28/2023] [Accepted: 09/13/2023] [Indexed: 12/24/2023] Open
Abstract
PURPOSE/OBJECTIVES Most patients with pancreatic adenocarcinoma (PDAC) will present with distant metastatic disease at diagnosis. We sought to identify clinical characteristics associated with prolonged overall survival (OS) in patients presenting with metastatic PDAC. MATERIALS/METHODS Patients presenting with metastatic PDAC that received treatment at our institution with FOLFIRINOX or gemcitabine-based chemotherapies between August 1, 2011 and September 1, 2017 were included in the study. Metastatic disease burden was comprehensively characterized radiologically via individual diagnostic imaging segmentation. Landmark analysis was performed at 18 months, and survival curves were estimated using the Kaplan-Meier method and compared between groups via the log-rank test. ECOG and Charlson Comorbidity Index (CCI) were calculated for all patients. RESULTS 121 patients were included with a median age of 62 years (37-86), 40% were female, 25% had ECOG 0 at presentation. Of the 121 patients included, 33% (n = 41) were alive at 12 months and 25% (n = 31) were alive at 18 months. Landmark analysis demonstrated a significant difference between patients surviving <18 months and ≥18 months regarding the presence of lung only metastases (36% vs. 16%, p = 0.04), number of organs with metastases (≥2 vs. 1, p = 0.04), and disease volume (mean of 19.1 cc vs. 1.4 cc, p = 0.04). At Year 1, predictors for improved OS included ECOG status at diagnosis (ECOG 0 vs. ECOG 1, p = 0.04), metastatic disease volume at diagnosis (≤0.1 cc vs. >60 cc, p = 0.004), metastasis only in the liver (p = 0.04), and normalization of CA 19-9 (p < 0.001). At Year 2, the only predictor of improved OS was normalization of the CA 19-9 (p = 0.03). In those patients that normalized their CA 19-9, median overall survival was 16 months. CONCLUSIONS In this exploratory analysis normalization of CA-19-9 or volumetric metastatic disease burden less than 0.2 cc demonstrated a remarkable OS, similar to that of patients with non-metastatic disease. These metrics are useful for counseling patients and identifying cohorts that may be optimal for trials exploring metastatic and/or local tumor-directed interventions.
Collapse
Affiliation(s)
| | - Alexis Visotcky
- Division of BiostatisticsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Anjishnu Banerjee
- Division of BiostatisticsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Mohammed Aldakkak
- Department of SurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - Susan Tsai
- Department of SurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - Douglas B. Evans
- Department of SurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - Kathleen K. Christians
- Department of SurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - Callisia N. Clarke
- Department of SurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - Ben George
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Aditya Shreenivas
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Mandana Kamgar
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Sakti Chakrabarti
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Kulwinder S. Dua
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Division of GastroenterologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Abdul Haq Khan
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Division of GastroenterologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Srivats Madhavan
- Division of Medical OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Division of GastroenterologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Beth A. Erickson
- Department of Radiation OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| | - William A. Hall
- Department of Radiation OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
- LaBahn Pancreatic Cancer ProgramMilwaukeeWisconsinUSA
| |
Collapse
|
590
|
Orlandi E, Citterio C, Chinelli R, Dotti I, Zaffignani E, Biasini C, Anselmi E, Cremona G, Vecchia S. Comparing Combination vs Monochemotherapy in Late-Elderly Patients With Advanced Pancreatic Cancer: Insights From a Single-Center Study. Cancer Control 2024; 31:10732748241304968. [PMID: 39620663 PMCID: PMC11613287 DOI: 10.1177/10732748241304968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES The incidence of pancreatic ductal adenocarcinoma (PDAC) increases with age and is frequently diagnosed at an inoperable stage, which limits treatment options. There is limited evidence concerning patients over 75 years old, and clinical practice often lacks clear guidance regarding the choice of first-line therapy. The primary objective of this retrospective study was to assess overall survival (OS) in elderly patients receiving first-line monochemotherapy vs combination therapy. Secondary objectives included evaluating progression-free survival (PFS) and safety. METHODS This retrospective study analyzed the records of 150 patients aged 75 or older with confirmed PDAC treated with first-line chemotherapy at Piacenza General Hospital, Italy. Of these, 72 patients received monotherapy, while 78 underwent polychemotherapy. The majority of patients (93.3%) were administered reduced doses, and within this group, 67.9% had their doses reduced by more than 20%. Most patients (80%) presented with comorbidities, predominantly hypertension and diabetes. RESULTS The median overall survival was significantly higher in the polychemotherapy group (8.2 months) compared to the monotherapy group (4.7 months), with a P-value of 0.0022. The median PFS was 5.7 months for polychemotherapy and 2.8 months for monotherapy, showing a statistically significant difference (P = 0.004). In the multivariate analysis, poor performance status, high CA19.9 levels, and monotherapy were significantly associated with worse OS. Patients treated with polychemotherapy had a 37% lower likelihood of death within the year compared to those treated with monotherapy (HR 0.58, P = 0.009). CONCLUSION Polychemotherapy may provide a survival advantage over monotherapy in the late-elderly population, although considerations for dose adjustments due to comorbidities and polypharmacy are necessary. These findings suggest that, when feasible, polychemotherapy could offer a balance between effectiveness and tolerability, potentially improving outcomes in this age group.
Collapse
Affiliation(s)
- Elena Orlandi
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Chiara Citterio
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Rebecca Chinelli
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Ilaria Dotti
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Elena Zaffignani
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Claudia Biasini
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Elisa Anselmi
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Gabriele Cremona
- Department of Oncology and Hematology, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Stefano Vecchia
- Department of Pharmacy, AUSL Piacenza Guglielmo da Saliceto Hospital, Piacenza, Italy
| |
Collapse
|
591
|
Igarashi T, Fukasawa M, Watanabe T, Kimura N, Itoh A, Tanaka H, Shibuya K, Yoshioka I, Hirabayashi K, Fujii T. Evaluating staging laparoscopy indications for pancreatic cancer based on resectability classification and treatment strategies for patients with positive peritoneal washing cytology. Ann Gastroenterol Surg 2024; 8:124-132. [PMID: 38250680 PMCID: PMC10797817 DOI: 10.1002/ags3.12719] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/17/2023] [Accepted: 07/04/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction The prognosis of pancreatic ductal adenocarcinoma (PDAC) in patients with positive peritoneal washing cytology (CY1) is poor. We aimed to evaluate the results of staging laparoscopy (SL) and treatment efficacy in CY1 patients based on a resectability classification. Methods We retrospectively reviewed 250 patients with PDAC who underwent SL before the initial treatment between 2017 and 2023 at the University of Toyama. Results The breakdown of cases by resectability classification was resectable (R):borderline resectable (BR):unresectable locally advanced (UR-LA) = 131:48:71 cases. The frequency of CY1 increased in proportion to the degree of local progression (R:BR:UR-LA = 20:23:34%), but the frequencies of liver metastasis or peritoneal dissemination were comparable (R:BR:UR-LA = 6.9:6.3:8.5%). Most CY1 patients received gemcitabine along with nab-paclitaxel therapy. The CY-negative conversion rates (R:BR:UR-LA = 70:64:52%) and conversion surgery rates (R:BR:UR-LA = 40:27:9%) were inversely proportional to the degree of local progression.Comparing H0P0CY1 factors for each classification, patients with H0P0CY1 had significantly more pancreatic body or tail carcinoma and tumor size ≥32 mm in R patients, whereas in BR patients, duke pancreatic monoclonal antigen type 2 (DUPAN-2) ≥ 230 U/mL was a significant factor. In contrast, no significant factors were observed in UR-LA patients. Conclusion The CY1 rates, CY-negative conversion rates, and conversion surgery rates varied according to local progression. In the case of R and BR, SL could be considered in patients with pancreatic body or tail carcinoma, large tumor size, or high DUPAN-2 level. In UR-LA, SL might be considered for all patients.
Collapse
Affiliation(s)
- Takamichi Igarashi
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Mina Fukasawa
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Toru Watanabe
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Nana Kimura
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Ayaka Itoh
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Haruyoshi Tanaka
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Kazuto Shibuya
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Isaku Yoshioka
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Kenichi Hirabayashi
- Department of Diagnostic Pathology, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of MedicineAcademic Assembly, University of ToyamaToyamaJapan
| |
Collapse
|
592
|
Prajapati R, Ostwal V, Srinivas S, Engineer R, Bhargava P, Saklani A, D'Souza A, Kumar S, Peelay Z, Manali P, Ramaswamy A. Modified FOLFIRINOX (mFOLFIRINOX) as neoadjuvant therapy and 'salvage' in patients with high risk locally advanced rectal cancers - tolerance and early outcomes. J Cancer Res Ther 2024; 20:199-203. [PMID: 38554321 DOI: 10.4103/jcrt.jcrt_225_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 09/15/2022] [Indexed: 04/01/2024]
Abstract
BACKGROUND There is limited data with regard to the use of modified 5-fluoroural-leucovorin-irinotecan-oxaliplatin (mFOLFIRINOX) in terms of tolerance and enabling total mesorectal excision (TME) of locally advanced rectal adenocarcinomas (LARC) with high-risk characteristics (T4b status, signet ring histology etc) post standard neoadjuvant long course chemoradiation (NACTRT) or short course radiation (SCRT) and chemotherapy. MATERIALS AND METHODS Patients with LARC from January 2018 to December 2020 receiving mFOLFIRINOX post NACTRT/SCRT to facilitate TME were evaluated. The primary endpoint was assessment of grade 3 and grade 4 treatment related toxicity and TME rates. Event free survival (EFS), where event was defined as disease progression or recurrence post resection after mFOLFIRINOX, was calculated by Kaplan Meier method. RESULTS Forty-seven patients were evaluated with a median age of 33 years (Range:18-59), 45% T4b status, 96% radiological circumferential margin (CRM) involved (79% CRM positive post NACTRT/SCRT), 43% extramural venous invasion (n=33) and 36% signet ring histology. 62% had received prior NACTRT and 38% had received SCRT with chemotherapy before receiving mFOLFIRINOX. The most common grade 3 and grade 4 treatment related side effects included diarrhoea (7%), anaemia (4%) and infections (4%). Intended duration of mFOLFIRINOX or beyond was completed in 94% of patients. 60% of patients underwent curative local resection with R0 resection rates of 100% (n=28) and pathological complete response rates of 21%. The most common surgeries done were exenterations and abdominoperineal in 22% and 17% patients respectively. With a median follow up of 19 months, 24 patients had recurred or progressed for a median EFS of 20 months [95% confidence interval (CI): 15-24]. CONCLUSIONS Locally advanced rectal cancers with high-risk characteristics are a niche group of cancers with less-than-optimal outcomes post standard neoadjuvant strategies. mFOLFIRINOX appears to be well tolerated and enables TME in a significant proportion of these patients.
Collapse
Affiliation(s)
- Ramjas Prajapati
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Reena Engineer
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Avanish Saklani
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Ashwin D'Souza
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Suman Kumar
- Department of Radiology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Zoya Peelay
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - P Manali
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| |
Collapse
|
593
|
Jo JH, Kim YT, Choi HS, Kim HG, Lee HS, Choi YW, Kim DU, Lee KH, Kim EJ, Han JH, Lee SO, Park CH, Choi EK, Kim JW, Cho JY, Lee WJ, Moon HR, Park MS, Kim S, Song SY. Efficacy of GV1001 with gemcitabine/capecitabine in previously untreated patients with advanced pancreatic ductal adenocarcinoma having high serum eotaxin levels (KG4/2015): an open-label, randomised, Phase 3 trial. Br J Cancer 2024; 130:43-52. [PMID: 37903909 PMCID: PMC10781743 DOI: 10.1038/s41416-023-02474-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The TeloVac study indicated GV1001 did not improve the survival of advanced pancreatic ductal adenocarcinoma (PDAC). However, the cytokine examinations suggested that high serum eotaxin levels may predict responses to GV1001. This Phase III trial assessed the efficacy of GV1001 with gemcitabine/capecitabine for eotaxin-high patients with untreated advanced PDAC. METHODS Patients recruited from 16 hospitals received gemcitabine (1000 mg/m2, D 1, 8, and 15)/capecitabine (830 mg/m2 BID for 21 days) per month either with (GV1001 group) or without (control group) GV1001 (0.56 mg; D 1, 3, and 5, once on week 2-4, 6, then monthly thereafter) at random in a 1:1 ratio. The primary endpoint was overall survival (OS) and secondary end points included time to progression (TTP), objective response rate, and safety. RESULTS Total 148 patients were randomly assigned to the GV1001 (n = 75) and control groups (n = 73). The GV1001 group showed improved median OS (11.3 vs. 7.5 months, P = 0.021) and TTP (7.3 vs. 4.5 months, P = 0.021) compared to the control group. Grade >3 adverse events were reported in 77.3% and 73.1% in the GV1001 and control groups (P = 0.562), respectively. CONCLUSIONS GV1001 plus gemcitabine/capecitabine improved OS and TTP compared to gemcitabine/capecitabine alone in eotaxin-high patients with advanced PDAC. CLINICAL TRIAL REGISTRATION NCT02854072.
Collapse
Affiliation(s)
- Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ho Soon Choi
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ho Gak Kim
- Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hong Sik Lee
- Department of Gastroenterology, Korea University College of Medicine, Seoul, Korea
| | - Young Woo Choi
- Department of Internal Medicine, Konyang University College of Medicine, Daejeon, Korea
| | - Dong Uk Kim
- Division of Gastroenterology and Hepatology, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Kwang Hyuck Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eui Joo Kim
- Division of Gastroenterology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Joung-Ho Han
- Department of Internal Medicine, Chungbuk National University College of Medicine & Chungbuk National University Hospital, Cheongju, South Korea
| | - Seung Ok Lee
- Department of Internal Medicine, The Research Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Korea
| | - Chang-Hwan Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Eun Kwang Choi
- Division of Gastroenterology, Department of Internal Medicine, Jeju National University College of Medicine, Jeju, Korea
| | - Jae Woo Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jae Yong Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
| | - Hyungsik Roger Moon
- Department of Economics, University of Southern California, Los Angeles, CA, USA
- Department of Economics, Yonsei University, Seoul, Korea
| | - Mi-Suk Park
- Department of Radiology, Yonsei University College of Medicine, Severance Hospital, Seoul, Korea
| | - Sangjae Kim
- GemVax & KAEL Co., Ltd. 58, Techno 11-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
594
|
Marcinak CT, Schwartz PB, Basree MM, Hurst N, Bassetti M, Kratz JD, Uboha NV. Treatment of Oligometastatic GI Cancers. Am Soc Clin Oncol Educ Book 2024; 44:e430152. [PMID: 38190577 DOI: 10.1200/edbk_430152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Oligometastatic state is believed to potentially represent a transitional stage between early, locoregional state disease and widely metastatic disease. Historically, locoregional approaches, particularly in advanced colorectal cancers, have demonstrated efficacy in select patients with limited burden of metastatic disease. Recent strides in systemic therapies, including biomarker-based treatments and immunotherapy, alongside innovations in surgical techniques and novel locoregional approaches such as stereotactic radiotherapy and ablation, have ushered in a new era of therapeutic possibilities across all oligometastatic GI cancers. Despite these advancements, there remains a significant gap in high-quality prospective evidence guiding patient selection and treatment decisions across various disease types. Ongoing clinical trials are anticipated to provide crucial insights into oligometastatic states, fostering the refinement of disease-specific oligometastatic state definitions and treatment algorithms. This article reviews existing data on the management of oligometastatic GI cancer, summarizes current state of knowledge for each disease state, and provides updates on ongoing studies in this space.
Collapse
Affiliation(s)
- Clayton T Marcinak
- Department of Surgery, University of Wisconsin School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI
| | - Patrick B Schwartz
- Department of Surgery, University of Wisconsin School of Medicine and Public Health University of Wisconsin-Madison, Madison, WI
| | - Mustafa M Basree
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Newton Hurst
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Michael Bassetti
- Department of Human Oncology, University of Wisconsin Hospital and Clinics, Madison, WI
| | - Jeremy D Kratz
- University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
- Center for Human Genomics and Precision Medicine, University of Wisconsin, Madison, WI
- William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Nataliya V Uboha
- University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| |
Collapse
|
595
|
Alonso-González R, Abadal Villayandre JM, Gálvez Gonzalez E, Álvarez Perez MJ, Méndez Alonso S, de Gregorio Ariza MA. Irreversible electroporation: Beyond the limits of tumor ablation. RADIOLOGIA 2024; 66:47-56. [PMID: 38365354 DOI: 10.1016/j.rxeng.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/02/2023] [Indexed: 02/18/2024]
Abstract
Irreversible Electroporation (IRE) is a non-thermal tumor ablation technique. High-voltage electrical pulses are applied between pairs of electrodes inserted around and/or inside a tumor. The generated electric current induces the creation of nanopores in the cell membrane, triggering apoptosis. As a result, IRE can be safely used in areas near delicate vascular structures where other thermal ablation methods are contraindicated. Currently, IRE has demonstrated to be a successful ablation technique for pancreatic, renal, and liver tumors and is widely used as a focal therapeutic option for prostate cancer. The need for specific anesthetic management and accurate parallel placement of multiple electrodes entails a high level of complexity and great expertise from the interventional team is required. Nevertheless, IRE is a very promising technique with a remarkable systemic immunological capability and may impact on distant metastases (abscopal effect).
Collapse
Affiliation(s)
- R Alonso-González
- Radiología Vascular Intervencionista, Hospital Universitario Severo Ochoa, Madrid, Spain.
| | - J M Abadal Villayandre
- Radiología Vascular Intervencionista, Hospital Universitario Severo Ochoa, Madrid, Spain
| | - E Gálvez Gonzalez
- Radiología Vascular Intervencionista, Hospital Universitario Severo Ochoa, Madrid, Spain
| | - M J Álvarez Perez
- Radiología Vascular Intervencionista, Hospital Universitario Severo Ochoa, Madrid, Spain
| | - S Méndez Alonso
- Radiología Vascular Intervencionista, Hospital Universitario Puerta Hierro, Madrid, Spain
| | | |
Collapse
|
596
|
de Castilhos J, Tillmanns K, Blessing J, Laraño A, Borisov V, Stein-Thoeringer CK. Microbiome and pancreatic cancer: time to think about chemotherapy. Gut Microbes 2024; 16:2374596. [PMID: 39024520 PMCID: PMC11259062 DOI: 10.1080/19490976.2024.2374596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by late diagnosis, rapid progression, and a high mortality rate. Its complex biology, characterized by a dense, stromal tumor environment with an immunosuppressive milieu, contributes to resistance against standard treatments like chemotherapy and radiation. This comprehensive review explores the dynamic role of the microbiome in modulating chemotherapy efficacy and outcomes in PDAC. It delves into the microbiome's impact on drug metabolism and resistance, and the interaction between microbial elements, drugs, and human biology. We also highlight the significance of specific bacterial species and microbial enzymes in influencing drug action and the immune response in the tumor microenvironment. Cutting-edge methodologies, including artificial intelligence, low-biomass microbiome analysis and patient-derived organoid models, are discussed, offering insights into the nuanced interactions between microbes and cancer cells. The potential of microbiome-based interventions as adjuncts to conventional PDAC treatments are discussed, paving the way for personalized therapy approaches. This review synthesizes recent research to provide an in-depth understanding of how the microbiome affects chemotherapy efficacy. It focuses on elucidating key mechanisms and identifying existing knowledge gaps. Addressing these gaps is crucial for enhancing personalized medicine and refining cancer treatment strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Juliana de Castilhos
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Katharina Tillmanns
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Jana Blessing
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Arnelyn Laraño
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Vadim Borisov
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Christoph K. Stein-Thoeringer
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| |
Collapse
|
597
|
Adekolujo OS, Wahab A, Akanbi MO, Oyasiji T, Hrinczenko B, Alese OB. Isolated pulmonary metastases in pancreatic ductal adenocarcinoma: a review of current evidence. Cancer Biol Ther 2023; 24:2198479. [PMID: 37526431 PMCID: PMC10395259 DOI: 10.1080/15384047.2023.2198479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/24/2023] [Indexed: 08/02/2023] Open
Abstract
Despite recent advances in cancer therapeutics, pancreatic ductal adenocarcinoma (PDAC) remains a lethal disease with a 5-year overall survival of only 10%. Since either at or within a few months of diagnosis, most patients with PDAC will present with metastatic disease, a more individualized approach to select patients who may benefit from more aggressive therapy has been suggested. Although studies have reported improved survival in PDAC and isolated pulmonary metastasis (ISP) compared to extrapulmonary metastases, such findings remain controversial. Furthermore, the added benefit of pulmonary metastasectomy and other lung-directed therapies remains unclear. In this review, we discuss the metastatic pattern of PDAC, evaluate the available evidence in the literature for improved survival in PDAC and ISP, evaluate the evidence for the added benefit of pulmonary metastasectomy and other lung-directed therapies, identify prognostic factors for survival, discuss the biological basis for the reported improved survival and identify areas for further research.
Collapse
Affiliation(s)
- Orimisan Samuel Adekolujo
- Department of Medicine, Michigan State University, East Lansing, MI, USA
- Medical Oncology, Karmanos Cancer Institute at McLaren Greater Lansing, Lansing, MI, USA
| | - Ahsan Wahab
- Department of Medicine, Prattville Baptist Hospital, Prattville, AL, USA
| | - Maxwell Oluwole Akanbi
- Department of Medicine, Michigan State University, East Lansing, MI, USA
- Department of Medicine, McLaren Flint, Flint, MI, USA
| | - Tolutope Oyasiji
- Department of Oncology, Barbara Ann Karmanos Cancer Institute at McLaren Flint, Wayne State University, Flint, MI, USA
| | - Borys Hrinczenko
- Department of Medicine, Michigan State University, East Lansing, MI, USA
- Medical Oncology, Karmanos Cancer Institute at McLaren Greater Lansing, Lansing, MI, USA
| | - Olatunji Boladale Alese
- Department of Hematology & Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
598
|
Kolbeinsson HM, Chandana S, Wright GP, Chung M. Pancreatic Cancer: A Review of Current Treatment and Novel Therapies. J INVEST SURG 2023; 36:2129884. [PMID: 36191926 DOI: 10.1080/08941939.2022.2129884] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Pancreatic cancer is one of the leading causes for cancer-related deaths in the United States. Majority of patients present with unresectable or metastatic disease. For those that present with localized disease, a multidisciplinary approach is necessary to maximize survival and optimize outcomes. The quality and safety of surgery for pancreatic cancer have improved in recent years with increasing adoption of minimally invasive techniques and surgical adjuncts. Systemic chemotherapy has also evolved to impact survival. It is now increasingly being utilized in the neoadjuvant setting, often with concomitant radiation. Increased utilization of genomic testing in metastatic pancreatic cancer has led to better understanding of their biology, thereby allowing clinicians to consider potential targeted therapies. Similarly, targeted agents such as PARP inhibitors and immune checkpoint- inhibitors have emerged with promising results. In summary, pancreatic cancer remains a disease with poor long-term survival. However, recent developments have led to improved outcomes and have changed practice in the past decade. This review summarizes current practices in pancreatic cancer treatment and the milestones that brought us to where we are today, along with emerging therapies.
Collapse
Affiliation(s)
- Hordur Mar Kolbeinsson
- Spectrum Health General Surgery Residency, Grand Rapids, Michigan, USA.,Department of Surgery, Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA
| | - Sreenivasa Chandana
- Department of Surgery, Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA.,Cancer and Hematology Centers of Western Michigan, PC, Grand Rapids, Michigan, USA
| | - G Paul Wright
- Spectrum Health General Surgery Residency, Grand Rapids, Michigan, USA.,Department of Surgery, Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA.,Division of Surgical Oncology, Spectrum Health Medical Group, Grand Rapids, Michigan, USA
| | - Mathew Chung
- Spectrum Health General Surgery Residency, Grand Rapids, Michigan, USA.,Department of Surgery, Michigan State University College of Human Medicine, Grand Rapids, Michigan, USA.,Division of Surgical Oncology, Spectrum Health Medical Group, Grand Rapids, Michigan, USA
| |
Collapse
|
599
|
Pang J, Zhuang B, Zhang LM. A co-carrier for plasmid DNA and curcumin delivery to treat pancreatic cancer via dendritic poly(l-lysine) modified amylose. Int J Biol Macromol 2023; 253:127467. [PMID: 37863141 DOI: 10.1016/j.ijbiomac.2023.127467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/22/2023]
Abstract
Pancreatic cancer is one of the most lethal malignancies in the world and remains one of the leading causes of cancer related death. For its treatment, a lot of investigations have dealt not only with individual chemotherapy by using polymeric carriers to deliver anticancer drugs, but also with individual gene therapy by using polymeric carriers to deliver nucleic acids such as small interfering RNA (siRNA) and plasmid DNA. However, relatively few studies have been focused on the co-delivery of gene and anticancer drug by multifunctional polymeric carriers for its synergistic therapy. In this work, a DPLL-functionalized amylose (ADP) was prepared by the click reaction between azidized amylose and propargyl focal point poly(l-lysine) dendrons, and then used to co-deliver plasmid pIRES2-EGFP-TNFα and curcumin for pancreatic cancer treatment. Due to the internal hydrophobic cavity of amylose component, ADP could load efficiently curcumin with anticancer activity and showed a sustained release behavior. Moreover, the curcumin-loaded ADP could form colloidally stable nanocomplexes with plasmid DNA in aqueous system due to the existence of cationic poly(l-lysine) dendrons and exhibited high gene transfection efficiency. The in vitro and in vivo tests confirmed the effectiveness of using ADP to co-deliver plasmid pIRES2-EGFP-TNFα and curcumin for synergistic therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Jiadong Pang
- DSAPM Lab and PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; Medical Intelligence and Innovation Academy, South University of Science and Technology Hospital, Shenzhen 518000, China
| | - Baoxiong Zhuang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li-Ming Zhang
- DSAPM Lab and PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
600
|
Zhou K, Strunk H, Dimitrov D, Vidal-Jove J, Gonzalez-Carmona MA, Essler M, Jin C, Mei Z, Zhu H, Marinova M. US-guided high-intensity focused ultrasound in pancreatic cancer treatment: a consensus initiative between Chinese and European HIFU centers. Int J Hyperthermia 2023; 41:2295812. [PMID: 38159562 DOI: 10.1080/02656736.2023.2295812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Purpose: Ultrasound-guided high-intensity focused ultrasound (USgHIFU) represents a safe and effective non-invasive thermoablative technique for managing inoperable pancreatic cancer. This treatment method significantly alleviates disease-related symptoms and reduces pancreatic tumor volume. However, the current body of evidence is constrained by a lack of randomized controlled trials. The utilization of USgHIFU is primarily indicated for patients with unresectable, locally advanced, or metastatic pancreatic cancer, particularly those experiencing symptoms due to a locally advanced primary tumor.Methods: This collaborative consensus paper, involving European and Chinese HIFU centers treating pancreatic cancer, delineates criteria for patient selection, focusing on those most likely to benefit from USgHIFU treatment. Consideration is given to endpoints encompassing symptom alleviation, local response rates, other oncological outcomes, as well as overall and progression-free survival. Additionally, this paper defines relevant contraindications, side effects, and complications associated with USgHIFU. The publication also explores the feasibility and role of USgHIFU within the context of palliative care, including standard systemic chemotherapy.Results: The non-invasive local treatment of advanced pancreatic cancer using HIFU should be regarded as an adjunctive option alongside systemic chemotherapy or best supportive care for managing this aggressive disease. Based on the ability of USgHIFU therapy to mitigate pain and reduce primary tumor volume, it should be considered as a complementary therapy for symptomatic patients with inoperable pancreatic cancer and as a potential means of tumor debulking. The underutilized yet promising USgHIFU exhibits the potential to enhance patients' quality of life by alleviating cancer-related pain. Experts in the field should evaluate this treatment option be evaluated by experts in this field, with this consensus paper potentially serving as a guiding resource for the medical community.Conclusions: US-guided HIFU for advanced pancreatic cancer addresses treatment goals, available options, success rates, and limitations. As a non-invasive, effective local therapy, complementary to chemotherapy and best supportive care, it plays a pivotal role in pain relief, reducing of tumor volume, and potentially improving survival rates.
Collapse
Affiliation(s)
- Kun Zhou
- Clinical Center for Tumor Therapy, 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | - Dobromir Dimitrov
- Department of Surgical Propedeutics, HIFU Center University Hospital St. Marina, Medical University Peleven, Pleven, Bulgaria
| | - Joan Vidal-Jove
- Institute Khuab, Comprehensive Tumor Center Barcelona, Barcelona, Spain
| | | | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Germany
| | - Chengbin Jin
- Clinical Center for Tumor Therapy, 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhechuan Mei
- Department of Gastroenterology and Hepatology, 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Zhu
- Clinical Center for Tumor Therapy, 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Milka Marinova
- Department of Nuclear Medicine, University Hospital Bonn, Germany
| |
Collapse
|