1
|
Hamed AB, El Asmar R, Tirukkovalur N, Tcharni A, Tatsuoka C, Jelinek M, Derby J, Dubrovsky G, Nunns G, Ongchin M, Pingpank JF, Zureikat AH, Bartlett DL, Singhi A, Choudry MH, AlMasri SS. Insight into Predictors of Cytoreduction Score Following Cytoreductive Surgery-Hyperthermic Intraperitoneal Chemotherapy for Gastric Peritoneal Carcinomatosis Improves Patient Selection and Prognostic Outcomes. Ann Surg Oncol 2025; 32:199-208. [PMID: 39382749 DOI: 10.1245/s10434-024-16328-z] [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: 02/20/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Peritoneal metastases due to gastric adenocarcinoma (GCPM) carry a dismal prognosis. A promising treatment strategy is cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC), but clear eligibility criteria for GCPM are lacking. We sought to identify factors associated with overall survival (OS) following CRS-HIPEC for GCPM to help optimize patient selection and clinical outcomes. PATIENTS AND METHODS In this single-center retrospective cohort study, we examined CRS-HIPEC outcomes for patients with GCPM between 2001 and 2021. After analyzing patient demographic, clinicopathologic, and perioperative variables, we applied multivariable Cox hazard models to assess factors associated with OS. We then assessed associations between baseline predictors and prognostically important variables using multivariable logistic regression. RESULTS We analyzed 55 patients with GCPM who underwent CRS-HIPEC. Median age was 54 years and 42% were female. Median peritoneal carcinomatosis index (PCI) was 8, and 75% of patients achieved a cytoreduction completeness score (CC score) of 0. Median progression-free survival (PFS) was 6.9 months, and median OS was 14.1 months. On adjusted analysis, a CC score > 0 (HR 2.3, p = 0.02) was significantly associated with worse OS. A peritoneal carcinomatosis index greater than 13 (OR 52.6, p = 0.001) and fewer lymph nodes (especially < 18) resected with the primary tumor (OR 0.86, p = 0.042) in the metachronous setting were significantly associated with incomplete macroscopic cytoreduction (CC score > 0). CONCLUSIONS We demonstrated that PCI > 13 and primary lymph nodes harvested < 18 in metachronous tumors are associated with CC score > 0, which in turn portends a worse OS. Although these results warrant prospective validation, they provide insight into improved selection of patients with GCPM for CRS-HIPEC.
Collapse
Affiliation(s)
- Ahmed B Hamed
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Rudy El Asmar
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nikhil Tirukkovalur
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adam Tcharni
- Faculty of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Curtis Tatsuoka
- Division of Hematology-Oncology, Department of Medicine, Pittsburgh, PA, USA
| | - Mark Jelinek
- UPMC Hillman Cancer Center Biostatistics Facility, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Joshua Derby
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Genia Dubrovsky
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Geoffrey Nunns
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Melanie Ongchin
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James F Pingpank
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amer H Zureikat
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David L Bartlett
- Department of Surgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Aatur Singhi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - M Haroon Choudry
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Samer S AlMasri
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Mechanicsburg, PA, USA.
| |
Collapse
|
2
|
Chen Y, Zhou Y, Xiong H, Wei Z, Zhang D, Li S. ASO Author Reflections: Advancing Gastric Cancer Treatment Through HIPEC-Current Insights and Future Directions. Ann Surg Oncol 2025; 32:252-253. [PMID: 39467970 DOI: 10.1245/s10434-024-16372-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024]
Affiliation(s)
- Ying Chen
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, Jiangsu, China
| | - Yang Zhou
- General Surgery Department, Siyang County First Peoples's Hospital, Siyang, Jiangsu, China
| | - Huaping Xiong
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, Jiangsu, China
| | - Zhen Wei
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, Jiangsu, China
| | - Dong Zhang
- General Surgery Department, Siyang County First Peoples's Hospital, Siyang, Jiangsu, China
| | - Shoushan Li
- Oncology Department of Siyang Hospital of Traditional Chinese Medicine, Siyang, Jiangsu, China.
| |
Collapse
|
3
|
Boshier PR, Tekkis N, Baggaley A, Robb HD, Lafaurie G, Simkens G, Nilsson M, Hanna GB, Petty R. Outcomes of intraperitoneal chemotherapy for the treatment of gastric cancer with peritoneal metastasis: A comprehensive systematic review and meta-analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 51:109499. [PMID: 39644811 DOI: 10.1016/j.ejso.2024.109499] [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: 10/02/2024] [Revised: 11/04/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Peritoneal metastasis is common in gastric cancer and linked to poor survival. Treatment of peritoneal metastasis with intraperitoneal chemotherapy has become an accepted practice in some centres. This systematic review and meta-analysis intends to provide a comprehensive evaluation of published evidence for the use of intraperitoneal chemotherapy is gastric cancer patients with peritoneal metastasis. METHODS A systematic literature search for studies reporting the use of intraperitoneal chemotherapy for the treatment gastric cancer with macroscopic peritoneal metastasis was performed up until June 2024. Studies were not eligible for inclusion if they described the use of intraperitoneal chemotherapy solely as an adjunct to gastrectomy or cytoreductive surgery. Pooled- and meta-analysis was used to summarise study outcomes. RESULTS Fifty-three studies reporting the outcomes of 2446 gastric cancer patients who received intraperitoneal chemotherapy for the treatment of peritoneal metastasis, were included. Three principal methods of intraperitoneal chemotherapy administration were described: catheter based (normothermic) intraperitoneal chemotherapy (n = 28); pressurised intraperitoneal aerosolised chemotherapy (n = 14), and; hyperthermic intraperitoneal chemotherapy (n = 11). The proportion of patients with complete peritoneal disease regression after receiving intraperitoneal chemotherapy was 27 % (95%CI, 14-41). Median overall survival determined was 16.4 months (95%CI, 14.4-18.4). Meta-analysis of data from eight studies comparing combined intraperitoneal and systemic chemotherapy with systemic chemotherapy alone identified a survival benefit for patients receiving intraperitoneal chemotherapy (Hazard ratio 0.57 [95%CI, 0.48-0.67],P < 0.001). CONCLUSION Despite variation in published treatment approaches and a lack of evidence from well-designed clinical trials, intraperitoneal chemotherapy may be considered safe and in selected circumstances efficacious.
Collapse
Affiliation(s)
- Piers R Boshier
- Department of Surgery and Cancer, Imperial College London, London, UK.
| | - Nicholas Tekkis
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alice Baggaley
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Henry D Robb
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Geert Simkens
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Magnus Nilsson
- Division of Surgery and Oncology, CLINTEC, Karolinska Institutet, Stokholm, Sweden
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Russell Petty
- Tayside Medical Science Centre, University of Dundee, Dundee, UK
| |
Collapse
|
4
|
Liang Z, Wang P, Li Z, Li W, Ma Q. Au Nanorings/TiO 2 NPs@MXene-Based Metasurfaces with a Magnetic Mirror-Modulated ECL Strategy for Extracellular Vesicle Detection. Anal Chem 2024; 96:16443-16452. [PMID: 39347690 DOI: 10.1021/acs.analchem.4c04460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
A metasurface as an artificial electromagnetic structure can concentrate optical energy into nanometric volumes to strongly enhance the light-matter interaction, which has been becoming a powerful platform for optical sensing, nonlinear effects, and quantum optics. Herein, we developed a novel hybrid plasmonic-dielectric metasurface consisting of Au nanorings (Au NRs) and TiO2 nanoparticles derived from MXene (TiO2 NPs@MXene). The hybrid metasurface simultaneously benefited from the high near-field enhancement effect of plasmonic materials and the low loss of dielectric materials. Furthermore, the optical modulation efficiency of the hybrid metasurface can be regulated by a magnetic mirror configuration. The magnetic mirror acted like a mirror, confining the electrons to a limited region and increasing the density of the surface plasmon. Moreover, the electrochemiluminescence (ECL) of the Cu2BDC metal-organic framework (Cu2BDC-MOF) served as a light source for the Au NRs/TiO2 NPs@MXene metasurface. Due to the exceptional light manipulation capability of the hybrid metasurface and the coordination of the magnetic mirror, the isotropic ECL signal can be dynamically amplified and converted into polarized emission. Finally, a metasurface-regulated ECL (MECL)-based biosensor with a dual-positive membrane protein recognition strategy was developed for the accurate identification of gastric cancer-derived extracellular vesicles. The novel MECL research opened up a new route in the realization of dynamically tunable metasurfaces for optical sensing and novel nanophotonic devices.
Collapse
Affiliation(s)
- Zihui Liang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenyan Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
5
|
Feng QX, Zhu ZN, Li Q, Liu XS. Dual-energy CT quantitative parameters to evaluate occult peritoneal metastasis in advanced gastric cancer preoperatively. Abdom Radiol (NY) 2024; 49:3309-3318. [PMID: 38634880 DOI: 10.1007/s00261-024-04303-x] [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: 01/20/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE To explore whether dual-energy CT (DECT) quantitative parameters could provide analytic value for the diagnosis of patients with occult peritoneal metastasis (OPM) in advanced gastric cancer preoperatively. MATERIALS AND METHODS This retrospective study included 219 patients with advanced gastric cancer and DECT scans. The patient's clinical data and DECT related iodine concentration (IC) parameters and effective atomic number (Zeff) were collated and analyzed among noun-peritoneal metastasis (NPM), OPM and radiologically peritoneal metastasis (RPM) groups. The predictive performance of the DECT parameters was compared with that of the conventional CT features and clinical characteristics through evaluating area under curve of the precision-recall (AUC-PR), F1 score, balanced accuracy, sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV). RESULTS Borrmann IV type diagnosed on CT and serum tumor indicator CA125 index were statistically different between the NPM and OPM groups. DECT parameters included IC, normalized IC (NIC), and Zeff of PM group were lower than the NPM group. The DECT predictive nomogram combined three independent DECT parameters produced a better diagnostic performance than the conventional CT feature Borrmann IV type and serum CA125 index in AUC-PR with 0.884 vs 0.368 vs 0.189, but similar to the combined indicator which was based on the DECT parameters, the conventional CT feature, and serum CA125 index in AUC-PR with 0.884 vs 0.918. CONCLUSION The lower quantitative NIC, IC ratio, and Zeff on DECT was associated with peritoneal metastasis in advanced gastric cancer and was promising to identify patients with OPM noninvasively.
Collapse
Affiliation(s)
- Qiu-Xia Feng
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Zhen-Ning Zhu
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Qiong Li
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China
| | - Xi-Sheng Liu
- Department of Radiology, The First Affiliated Hospital With Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
6
|
van der Sluis K, Vollebergh MA, Kodach LL, van Dieren JM, de Hingh IHJT, Wijnhoven BPL, Verhoeven RHA, Sandick JWV. The clinical implications of staging laparoscopy in the diagnostic workup of gastric cancer patients: A population based study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108569. [PMID: 39134081 DOI: 10.1016/j.ejso.2024.108569] [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: 04/10/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Since 2016, staging laparoscopy has been implemented in the diagnostic workup of patients with gastric cancer. Staging laparoscopy aims to detect incurable disease (peritoneal metastases and irresectable tumors) and to prevent futile laparotomies. METHODS In this population-based nationwide study, we sought patient- and tumor characteristics associated with undergoing a staging laparoscopy. Additionally, we analyzed the prevalence of synchronous peritoneal metastases, the outcome of the staging laparoscopy and its clinical impact on treatment decisions. All patients diagnosed with non-cardia gastric cancer from the Netherlands Cancer Registry between 2016 and 2021 were included. RESULTS Alongside tumor characteristics, patient characteristics such as younger age, absence of comorbidities and lower WHO performance status were associated with performing a staging laparoscopy. In the study period, an increase in the proportion of patients who underwent a staging laparoscopy was observed, from 19.6% in 2016 to 32.3% in 2021 (p-value<0.001). In the same period, the prevalence of synchronous peritoneal metastases increased from 25% to 31%. In 37.6% of the patients who had the outcome of their staging laparoscopy reported, had incurable disease diagnosed during staging laparoscopy. Significantly less of these patients were treated with triplet regimens as compared to patients with a negative staging laparoscopy (18.5 vs. 76.3%; p-value<0.001). CONCLUSION The implementation of staging laparoscopy in gastric cancer patients paralleled the increase in diagnosis of incurable disease and a decrease in the application of triplet systemic therapies in these patients.
Collapse
Affiliation(s)
- K van der Sluis
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands.
| | - M A Vollebergh
- The Netherlands Cancer Institute, Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - L L Kodach
- The Netherlands Cancer Institute, Department of Pathology, Amsterdam, the Netherlands
| | - J M van Dieren
- The Netherlands Cancer Institute, Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - I H J T de Hingh
- Catharina Hospital, Department of Surgery, Eindhoven, the Netherlands; Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands; Netherlands Comprehensive Cancer Organization (IKNL), Department of Research & Development, Utrecht, the Netherlands
| | - B P L Wijnhoven
- Erasmus Medical Centre, Department of Surgery, Rotterdam, the Netherlands
| | - R H A Verhoeven
- Netherlands Comprehensive Cancer Organization (IKNL), Department of Research & Development, Utrecht, the Netherlands; Amsterdam UMC Location University of Amsterdam, Medical Oncology, Meibergdreef 9, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - J W van Sandick
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Zhang Y, Xie J. Targeting non-coding RNAs as a promising biomarker in peritoneal metastasis: Background, mechanism, and therapeutic approach. Biomed Pharmacother 2024; 179:117294. [PMID: 39226726 DOI: 10.1016/j.biopha.2024.117294] [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: 06/09/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/05/2024] Open
Abstract
Peritoneal metastasis (PM) pathophysiology is complex and not fully understood. PM, originating from gastrointestinal (GI) cancer, is a condition that significantly worsens patient prognosis due to its complex nature and limited treatment options. The non-coding RNAs (ncRNAs) have been shown to play pivotal roles in cancer biology, influencing tumorigenesis, progression, metastasis, and therapeutic resistance. Increasing evidence has demonstrated the regulatory functions of different classes of ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in PM. Identifying biomarkers for early detection of PM is a crucial step towards improving patient outcomes, and how ncRNA profiles correlate with survival rates, response to therapy, and recurrence risks have raised much attention in recent years. Additionally, exploring innovative therapeutic approaches utilizing ncRNAs, such as targeted therapy and gene silencing, may offer new horizons in treating this dire condition. Recent advances in systemic treatments and the development of novel loco-regional therapies have opened doors to multimodal treatment approaches. Radical surgeries combined with hyperthermic intraperitoneal chemotherapy (HIPEC) have shown promising results, leading to extended patient survival. Current research is focused on the molecular characterization of PM, which is crucial for early detection and developing future therapeutic strategies. By summarizing the latest findings, this study underscores the transformative potential of ncRNAs in enhancing the diagnosis, prognosis, and treatment of PM in GI cancer, paving the way for more personalized and effective clinical strategies.
Collapse
Affiliation(s)
- Yiping Zhang
- School of Life Sciences, Fudan University, Shanghai 200438, China; Wanchuanhui (Shanghai) Medical Technology Co., Ltd, Shanghai 201501, China.
| | - Jun Xie
- School of Life Sciences, Fudan University, Shanghai 200438, China; Wanchuanhui (Shanghai) Medical Technology Co., Ltd, Shanghai 201501, China.
| |
Collapse
|
8
|
Wu K, Qiu C, Ma Q, Chen F, Lu T. The anti-cancer mechanism of Celastrol by targeting JAK2/STAT3 signaling pathway in gastric and ovarian cancer. Toxicol Appl Pharmacol 2024; 491:117077. [PMID: 39181414 DOI: 10.1016/j.taap.2024.117077] [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: 06/21/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Celastrol is a natural triterpene exhibiting significant and extensive antitumor activity in a wide range of cancer. Due to unfavorable toxicity profile and undefined mechanism, Celastrol's application in clinical cancer therapy remains limited. Herein, we elucidate the pharmacological mechanism of Celastrol's anticancer effects, with a focus on STAT3 signaling pathway in cancers with high incidence of metastasis. METHODS The safety profile of Celastrol were assessed in mice. In vitro analysis was performed in gastric cancer and ovarian cancer to assess the cytotoxicity, induction of reactive oxygen species (ROS) of Celastrol using STAT3 knockout cancer cells. Effects of Celastrol on STAT3 activation and transcription activity, JAK2/STAT3 signaling protein expression were assessed. Additionally, proteomic contrastive analysis was performed to explore the molecular association of Celastrol with STAT3 deletion in cancer cells. RESULTS Celastrol has no obvious toxic effect at 1.5 mg/kg/day in a 15 days' administration. Celastrol inhibits tumor growth and increases ROS in a STAT3 dependent manner in gastric and ovarian cancer celllines. On molecular level, it downregulates IL-6 level and inhibits the JAK2/STAT3 signaling pathway by suppressing STAT3' activation and transcription activity. Proteomic contrastive analysis suggests a similar cellular mechanism of action between Celastrol and STAT3 deletion on regulating cancer progression pathways related to migration and invasion. CONCLUSION Our research elucidates the anti-cancer mechanism of Celastrol through targeting the JAK2/STAT3 signaling pathway in cancer with high incidence of metastasis. This study provides a solid theoretical basis for the application of Celastrol in cancer therapy.
Collapse
Affiliation(s)
- Kang Wu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chentao Qiu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qihong Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Fangfang Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tiangong Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
9
|
González-Moreno S, Deraco M, Kusamura S, Bijelic L, Lambert LA, Dromain C, Bibeau F, Liauw W, Reis A, Galan A, Sugarbaker PH. Education and Training in Peritoneal Surface Oncology. J Surg Oncol 2024. [PMID: 39318155 DOI: 10.1002/jso.27833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 09/26/2024]
Abstract
Peritoneal surface oncology (PSO) is a novel field dealing with the knowledge of peritoneal neoplasms, primary or secondary, and their clinical management. As a specific treatment with curative intent for peritoneal neoplasms developed over the years, there is a growing need to comprehensively educate and train surgical oncologists worldwide in this discipline, a recognized unmet need. The European School of Peritoneal Surface Oncology (ESPSO) emerged in 2014 to provide an answer to this need.
Collapse
Affiliation(s)
- Santiago González-Moreno
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- MD Anderson Cancer Center, Madrid, Spain
| | - Marcello Deraco
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Shigeki Kusamura
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lana Bijelic
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Consorci Hospitalari Moisés Broggi, Barcelona, Spain
| | - Laura A Lambert
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- University of Utah Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Clarisse Dromain
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Frédéric Bibeau
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Besançon University Hospital, Franche Comté University, Besançon, France
| | - Winston Liauw
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Cancer Care Centre, St George Hospital, University of New South Wales, Sidney, Australia
| | - Artur Reis
- Santa Casa de São José dos Campos, São Paulo, Brazil
| | - Ana Galan
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- European Society of Surgical Oncology, Brussels, Belgium
| | - Paul H Sugarbaker
- European School of Peritoneal Surface Oncology, Brussels, Belgium
- Washington Cancer Institute, Washington, DC, USA
| |
Collapse
|
10
|
Li D, Jin Y, He X, Deng J, Lu W, Yang Z, Zheng X, Hou K, Tang S, Bao B, Ren J, Zhang X, Wang J, Yan H, Qu X, Liu Y, Che X. Hypoxia-induced LAMB2-enriched extracellular vesicles promote peritoneal metastasis in gastric cancer via the ROCK1-CAV1-Rab11 axis. Oncogene 2024; 43:2768-2780. [PMID: 39138263 DOI: 10.1038/s41388-024-03124-y] [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: 11/26/2023] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Peritoneal metastasis is one of the most common risk factors contributing to the poor prognosis of gastric cancer. We previously reported that extracellular vesicles from gastric cancer cells could facilitate peritoneal metastasis. However, their impact on gastric cancer-induced peritoneal metastasis under hypoxic conditions remains unclear. This study aims to elucidate how hypoxia-resistant gastric cancer cell-derived extracellular vesicles affect the peritoneal metastasis of normoxic gastric cancer cells. Proteomic analysis revealed elevated levels of Caveolin1 and Laminin β2 in hypoxia-resistant gastric cancer cells and their corresponding extracellular vesicles. Importantly, Caveolin1 was found to play a central role in mediating Laminin β2 sorting into extracellular vesicles derived from hypoxia-resistant gastric cancer cells, and subsequently, extracellular vesicle-associated Laminin β2 promoted peritoneal metastasis in normoxic gastric cancer cells by activating the AKT pathway. Further investigation confirmed that Caveolin1 activation by Rho-related Coiled-coil kinase 1-mediated phosphorylation of Y14 residue is a key factor facilitating Laminin β2 sorting into extracellular vesicles. Moreover, Y14 phosphorylated- Caveolin1 enhanced Laminin β2 sorting by activating Rab11. Finally, our study demonstrated that a combined assessment of plasma extracellular vesicle-associated Caveolin1 and extracellular vesicle-associated Laminin β2 could provide an accurate predictive tool for peritoneal metastasis occurrence in gastric cancer.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Yue Jin
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xin He
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Jian Deng
- Third Department of Medical Oncology, The Fifth People Hospital of Shenyang, Shenyang, China
| | - Wenqing Lu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Zichang Yang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xueying Zheng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Shiying Tang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Bowen Bao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Jie Ren
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xiaojie Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Jin Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Hongfei Yan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Research Center of Shenyang, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
- Clinical Cancer Research Center of Shenyang, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China.
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
- Clinical Cancer Research Center of Shenyang, Shenyang, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China.
| |
Collapse
|
11
|
Bohosova J, Ashraf NS, Slaby O, Calin GA. Non-Coding RNAs in Peritoneal Carcinomatosis: From Bench to Bedside. Cancers (Basel) 2024; 16:2961. [PMID: 39272819 PMCID: PMC11394633 DOI: 10.3390/cancers16172961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Peritoneal carcinomatosis represents an advanced stage of tumors within the peritoneal cavity. Once considered an incurable terminal cancer metastasis, contemporary medicine is on the hunt for certain potentially curative options alongside the present day's palliative disease management. However, for most patients, peritoneal carcinomatosis continues to pose a fatal late-stage prognosis with a grim future outlook. Over the past two decades, non-coding RNAs have garnered significant attention due to their undeniable significance in regulating cellular processes across all levels. Disruption of the intricate regulation led by non-coding RNAs has been demonstrated to have a substantial impact on various human diseases, particularly in cancer, including solid tumors originating from the organs of the peritoneal cavity. This review aims to offer a comprehensive overview of the current state of knowledge in the under-researched field of peritoneal carcinomatosis, focusing specifically on the role of non-coding RNAs in the development of this condition and delineating potential avenues for future research.
Collapse
Affiliation(s)
- Julia Bohosova
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Nida Sarosh Ashraf
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Islamabad 45550, Pakistan
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - George A Calin
- Translational Molecular Pathology Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The RNA Interference and Non-Coding RNA Center, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
12
|
Ramos C, Gerakopoulos V, Oehler R. Metastasis-associated fibroblasts in peritoneal surface malignancies. Br J Cancer 2024; 131:407-419. [PMID: 38783165 PMCID: PMC11300623 DOI: 10.1038/s41416-024-02717-4] [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: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Over decades, peritoneal surface malignancies (PSMs) have been associated with limited treatment options and poor prognosis. However, advancements in perioperative systemic chemotherapy, cytoreductive surgery (CRS), and hyperthermic intraperitoneal chemotherapy (HIPEC) have significantly improved clinical outcomes. PSMs predominantly result from the spread of intra-abdominal neoplasia, which then form secondary peritoneal metastases. Colorectal, ovarian, and gastric cancers are the most common contributors. Despite diverse primary origins, the uniqueness of the peritoneum microenvironment shapes the common features of PSMs. Peritoneal metastization involves complex interactions between tumour cells and the peritoneal microenvironment. Fibroblasts play a crucial role, contributing to tumour development, progression, and therapy resistance. Peritoneal metastasis-associated fibroblasts (MAFs) in PSMs exhibit high heterogeneity. Single-cell RNA sequencing technology has revealed that immune-regulatory cancer-associated fibroblasts (iCAFs) seem to be the most prevalent subtype in PSMs. In addition, other major subtypes as myofibroblastic CAFs (myCAFs) and matrix CAFs (mCAFs) were frequently observed across PSMs studies. Peritoneal MAFs are suggested to originate from mesothelial cells, submesothelial fibroblasts, pericytes, endothelial cells, and omental-resident cells. This plasticity and heterogeneity of CAFs contribute to the complex microenvironment in PSMs, impacting treatment responses. Understanding these interactions is crucial for developing targeted and local therapies to improve PSMs patient outcomes.
Collapse
Affiliation(s)
- Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Vasileios Gerakopoulos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
13
|
Pereira MA, Ramos MFKP, Charruf AZ, Dias AR, Ribeiro U. Risk Scoring System to Predict Mortality in Gastric Cancer with Peritoneal Carcinomatosis. Med Sci (Basel) 2024; 12:30. [PMID: 38921684 PMCID: PMC11205632 DOI: 10.3390/medsci12020030] [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: 04/18/2024] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Gastric cancer (GC) with peritoneal carcinomatosis (PC) has a particularly unfavorable prognosis. This limited survival raises doubts about which factors confer an extremely worse outcome and which patients could benefit from more aggressive treatments, in an attempt to improve survival and better control the disease. This study aimed to evaluate the survival outcomes of patients with PC due to GC and develop a prognostic score to predict 6-month mortality. We performed an analysis of clinical stage IV GC with PC. Scores were assigned to risk factors and calculated for each patient from nine variables. Among 326 IVB GC, 211 (64.7%) had PC and were included. After calculating the score, 136 (64.5%) GCs were classified as a low-risk group and 75 (35.5%) as a high-risk group. Median OS was 7.9 and 1.9 months for low- and high-risk patients (p < 0.001). In the high-risk group, 77.3% of the patients died in <6 mo (p < 0.001). Palliative surgery and chemotherapy were associated with better survival, and the prognostic groups maintained statistical significance even when the same type of treatment was performed. In conclusion, the scoring system developed with variables related to patient performance status and clinical data was able to distinguish GC with PC with a high risk of 6-month mortality. Accordingly, verifying and validating our findings in a large cohort of patients is necessary to confirm and guarantee the external validation of the results.
Collapse
Affiliation(s)
- Marina Alessandra Pereira
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01249-000, Brazil; (M.F.K.P.R.); (A.Z.C.); (A.R.D.)
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Av. Dr. Arnaldo, 251, São Paulo 01246-000, Brazil
| | - Marcus Fernando Kodama Pertille Ramos
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01249-000, Brazil; (M.F.K.P.R.); (A.Z.C.); (A.R.D.)
| | - Amir Zeide Charruf
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01249-000, Brazil; (M.F.K.P.R.); (A.Z.C.); (A.R.D.)
| | - André Roncon Dias
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01249-000, Brazil; (M.F.K.P.R.); (A.Z.C.); (A.R.D.)
| | - Ulysses Ribeiro
- Department of Gastroenterology, Instituto do Cancer, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01249-000, Brazil; (M.F.K.P.R.); (A.Z.C.); (A.R.D.)
| |
Collapse
|
14
|
Yu P, Ding G, Huang X, Wang C, Fang J, Huang L, Ye Z, Xu Q, Wu X, Yan J, Ou Q, Du Y, Cheng X. Genomic and immune microenvironment features influencing chemoimmunotherapy response in gastric cancer with peritoneal metastasis: a retrospective cohort study. Int J Surg 2024; 110:3504-3517. [PMID: 38502852 PMCID: PMC11175815 DOI: 10.1097/js9.0000000000001281] [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: 11/14/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Patients with peritoneal metastasis (PM) from gastric cancer (GC) exhibit poor prognosis. Chemoimmunotherapy offers promising clinical benefits; however, its efficacy and predictive biomarkers in a conversion therapy setting remain unclear. The authors aimed to retrospectively evaluate chemoimmunotherapy efficacy in a conversion therapy setting for GC patients with PM and establish a prediction model for assessing clinical benefits. MATERIALS AND METHODS A retrospective evaluation of clinical outcomes encompassed 55 GC patients with PM who underwent chemoimmunotherapy in a conversion therapy setting. Baseline PM specimens were collected for genomic and transcriptomic profiling. Clinicopathological factors, gene signatures, and tumor immune microenvironment were evaluated to identify predictive markers and develop a prediction model. RESULTS Chemoimmunotherapy achieved a 41.8% objective response rate and 72.4% R0 resection rate in GC patients with PM. Patients with conversion surgery showed better overall survival (OS) than those without the surgery (median OS: not reached vs 7.82 m, P <0.0001). Responders to chemoimmunotherapy showed higher ERBB2 and ERBB3 mutation frequencies, CTLA4 and HLA-DQB1 expression, and CD8+ T cell infiltration, but lower CDH1 mutation and naïve CD4+ T cell infiltration, compared to nonresponders. A prediction model was established integrating CDH1 and ERBB3 mutations, HLA-DQB1 expression, and naïve CD4+ T cell infiltration (AUC=0.918), which were further tested using an independent external cohort (AUC=0.785). CONCLUSION This exploratory study comprehensively evaluated clinicopathological, genomic, and immune features and developed a novel prediction model, providing a rational basis for the selection of GC patients with PM for chemoimmunotherapy-involved conversion therapy.
Collapse
Affiliation(s)
- Pengfei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Guangyu Ding
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Xingmao Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Chenxuan Wang
- Medical department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People’s Republic of China
| | - Jingquan Fang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Ling Huang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Zeyao Ye
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Qi Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang
| | - Xiaoying Wu
- Medical department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People’s Republic of China
| | - Junrong Yan
- Medical department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People’s Republic of China
| | - Qiuxiang Ou
- Medical department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, People’s Republic of China
| | - Yian Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences
| |
Collapse
|
15
|
Zhou J, Cai X, Lu Z, Xiong B, Peng C. Short-Term Safety Evaluation of Albumin-Bound Paclitaxel in Intraoperative and Postoperative Hyperthermic Intraperitoneal Chemotherapy for Gastric Cancer. J Gastrointest Cancer 2024; 55:877-887. [PMID: 38367177 DOI: 10.1007/s12029-024-01031-9] [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] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
PURPOSE To evaluate the short-term safety of albumin-bound paclitaxel in hyperthermic intraperitoneal chemotherapy (HIPEC) during and after gastric cancer (GC) surgery. METHODS A retrospective analysis of clinical data was conducted for GC surgery patients at Zhongnan Hospital of Wuhan University, from January 2020 to September 2022. The study group (n = 120) received HIPEC and the control group (n = 268) did not receive albumin-bound paclitaxel. Short-term safety indicators including intraoperative complications, hematological toxicity, liver and kidney function, and gastrointestinal function recovery were compared between the two groups. RESULTS There were no statistically significant differences between the two groups regarding intraoperative complications, hematological toxicity, liver and kidney function, and gastrointestinal function recovery time (P > 0.05 for all). In the study group, patients were further divided into subgroups based on dose and timing. Subgroup analysis revealed no significant differences among the different dose subgroups. However, when focusing on timing subgroups, the postoperative subgroup exhibited significantly higher white blood cell counts and bilirubin levels compared to the intraoperative subgroup, while the intraoperative subgroup had significantly higher bilirubin levels compared to both postoperative and intraoperative plus postoperative subgroups. CONCLUSION Albumin-bound paclitaxel demonstrates good safety and tolerability in HIPEC during and after GC surgery, without increasing the risk of intraoperative complications.
Collapse
Affiliation(s)
- Jingxiang Zhou
- Second Clinical College, Wuhan University, Wuhan , Hubei Province, 430070, China
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Xiaopeng Cai
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Zhao Lu
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China
| | - Chunwei Peng
- Department of Gastrointestinal Surgery, the Center for Minimally Invasive Surgery of Gastrointestinal Cancer, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan Peritoneal Cancer Clinical Medical Center, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Hubei Province, 430070, China.
| |
Collapse
|
16
|
Guchelaar NAD, Nasserinejad K, Mostert B, Koolen SLW, van der Sluis PC, Lagarde SM, Wijnhoven BPL, Mathijssen RHJ, Noordman BJ. Intraperitoneal chemotherapy for peritoneal metastases of gastric origin: a systematic review and meta-analysis. Br J Surg 2024; 111:znae116. [PMID: 38722803 PMCID: PMC11081074 DOI: 10.1093/bjs/znae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Gastric cancer with peritoneal metastases is associated with a dismal prognosis. Normothermic catheter-based intraperitoneal chemotherapy and normothermic pressurized intraperitoneal aerosol chemotherapy (PIPAC) are methods to deliver chemotherapy intraperitoneally leading to higher intraperitoneal concentrations of cytotoxic drugs compared to intravenous administration. We reviewed the effectiveness and safety of different methods of palliative intraperitoneal chemotherapy. METHODS Embase, MEDLINE, Web of Science and Cochrane were searched for articles studying the use of repeated administration of palliative intraperitoneal chemotherapy in patients with gastric cancer and peritoneal metastases, published up to January 2024. The primary outcome was overall survival. RESULTS Twenty-three studies were included, representing a total of 999 patients. The pooled median overall survival was 14.5 months. The pooled hazard ratio of the two RCTs using intraperitoneal paclitaxel and docetaxel favoured the intraperitoneal chemotherapy arm. The median overall survival of intraperitoneal paclitaxel, intraperitoneal docetaxel and PIPAC with cisplatin and doxorubicin were respectively 18.4 months, 13.2 months and 9.0 months. All treatment methods had a relatively safe toxicity profile. Conversion surgery after completion of intraperitoneal therapy was performed in 16% of the patients. CONCLUSIONS Repeated intraperitoneal chemotherapy, regardless of method of administration, is safe for patients with gastric cancer and peritoneal metastases. Conversion surgery after completion of the intraperitoneal chemotherapy is possible in a subset of patients.
Collapse
Affiliation(s)
- Niels A D Guchelaar
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Kazem Nasserinejad
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
- HOVON Foundation, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
- Department of Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Pieter C van der Sluis
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Sjoerd M Lagarde
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Bo J Noordman
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Pelc Z, Sędłak K, Leśniewska M, Mielniczek K, Chawrylak K, Skórzewska M, Ciszewski T, Czechowska J, Kiszczyńska A, Wijnhoven BPL, Van Sandick JW, Gockel I, Gisbertz SS, Piessen G, Eveno C, Bencivenga M, De Manzoni G, Baiocchi GL, Morgagni P, Rosati R, Fumagalli Romario U, Davies A, Endo Y, Pawlik TM, Roviello F, Bruns C, Polkowski WP, Rawicz-Pruszyński K. Textbook Neoadjuvant Outcome-Novel Composite Measure of Oncological Outcomes among Gastric Cancer Patients Undergoing Multimodal Treatment. Cancers (Basel) 2024; 16:1721. [PMID: 38730672 PMCID: PMC11083243 DOI: 10.3390/cancers16091721] [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: 04/11/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
The incidence of gastric cancer (GC) is expected to increase to 1.77 million cases by 2040. To improve treatment outcomes, GC patients are increasingly treated with neoadjuvant chemotherapy (NAC) prior to curative-intent resection. Although NAC enhances locoregional control and comprehensive patient care, survival rates remain poor, and further investigations should establish outcomes assessment of current clinical pathways. Individually assessed parameters have served as benchmarks for treatment quality in the past decades. The Outcome4Medicine Consensus Conference underscores the inadequacy of isolated metrics, leading to increased recognition and adoption of composite measures. One of the most simple and comprehensive is the "All or None" method, which refers to an approach where a specific set of criteria must be fulfilled for an individual to achieve the overall measure. This narrative review aims to present the rationale for the implementation of a novel composite measure, Textbook Neoadjuvant Outcome (TNO). TNO integrates five objective and well-established components: Treatment Toxicity, Laboratory Tests, Imaging, Time to Surgery, and Nutrition. It represents a desired, multidisciplinary care and hospitalization of GC patients undergoing NAC to identify the treatment- and patient-related data required to establish high-quality oncological care further. A key strength of this narrative review is the clinical feasibility and research background supporting the implementation of the first and novel composite measure representing the "ideal" and holistic care among patients with locally advanced esophago-gastric junction (EGJ) and GC in the preoperative period after NAC. Further analysis will correlate clinical outcomes with the prognostic factors evaluated within the TNO framework.
Collapse
Affiliation(s)
- Zuzanna Pelc
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Sędłak
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Magdalena Leśniewska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Mielniczek
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Katarzyna Chawrylak
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Magdalena Skórzewska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Tomasz Ciszewski
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Joanna Czechowska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Agata Kiszczyńska
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Bas P. L. Wijnhoven
- Department of General Surgery, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Johanna W. Van Sandick
- Department of Surgical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Suzanne S. Gisbertz
- Department of Surgery, Amsterdam UMC location University of Amsterdam, 1007 MB Amsterdam, The Netherlands;
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, University Lille, and Claude Huriez University Hospital, 59000 Lille, France; (G.P.); (C.E.)
| | - Clarisse Eveno
- Department of Digestive and Oncological Surgery, University Lille, and Claude Huriez University Hospital, 59000 Lille, France; (G.P.); (C.E.)
| | - Maria Bencivenga
- Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Giovanni De Manzoni
- Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, Surgical Clinic, University of Brescia, and Third Division of General Surgery, Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Paolo Morgagni
- Department of General Surgery, Morgagni-Pierantoni Hospital, 47121 Forlì, Italy;
| | - Riccardo Rosati
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Hospital, Vita Salute University, 20132 Milan, Italy;
| | | | - Andrew Davies
- Department of Upper Gastrointestinal and General Surgery, Guy’s and St Thomas’ Hospital, London SE1 7EH, UK;
| | - Yutaka Endo
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.E.); (T.M.P.)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.E.); (T.M.P.)
| | - Franco Roviello
- Department of Medicine, Surgery, and Neurosciences, University of Siena, 53100 Siena, Italy;
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, 50937 Cologne, Germany;
| | - Wojciech P. Polkowski
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| | - Karol Rawicz-Pruszyński
- Department of Surgical Oncology, Medical University of Lublin, 20079 Lublin, Poland; (Z.P.); (K.S.); (M.L.); (K.M.); (K.C.); (M.S.); (T.C.); (J.C.); (A.K.); (W.P.P.)
| |
Collapse
|
18
|
Zhang S, Hu Q, Chen X, Zhou N, Huang Q, Tan S, Su M, Gou H. 68Ga-FAPI-04 positron emission tomography/CT and laparoscopy for the diagnosis of occult peritoneal metastasis in newly diagnosed locally advanced gastric cancer: study protocol of a single-centre prospective cohort study. BMJ Open 2024; 14:e075680. [PMID: 38643004 PMCID: PMC11033661 DOI: 10.1136/bmjopen-2023-075680] [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: 05/15/2023] [Accepted: 03/22/2024] [Indexed: 04/22/2024] Open
Abstract
INTRODUCTION Accurate baseline clinical staging is critical to inform treatment decision-making for patients with gastric cancers. Peritoneal metastasis (PM) is the most common form of metastasis in gastric cancer and mainly diagnosed by diagnostic laparoscopy and peritoneal lavage evaluation. However, diagnostic laparoscopy is invasive and less cost-effective. It is urgent to develop a safe, fast and non-invasive functional imaging method to verify the peritoneal metastasis of gastric cancer. The aim of our study was to evaluate the proportion of patients in whom 68Ga-FAPI-04 positron emission tomography/CT (PET/CT) led to a change in treatment strategy and to assess the diagnostic accuracy of 68Ga-FAPI-04 PET/CT for the detection of occult peritoneal metastasis compared with laparoscopic exploration. METHODS AND ANALYSIS In this single-centre, prospective diagnostic test accuracy study, a total of 48 patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma (cT4a-b, N0-3, M0, based on CT images) who are considering radical tumour surgery will be recruited. All participants will undergo 68Ga-FAPI-04 PET/CT before the initiation of laparoscopic exploration. The primary outcome is the proportion of patients with occult peritoneal metastatic lesions detected by 68Ga-FAPI-04 PET/CT, leading to a change in therapy strategy. The secondary outcomes include the diagnostic performance of 68Ga-FAPI-04 PET/CT for occult peritoneal metastasis, including sensitivity, specificity, accuracy, positive predictive value and negative predictive value. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of West China Hospital, Sichuan University (2022-1484). Study results will be presented at public and scientific conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER ChiCTR2300067591.
Collapse
Affiliation(s)
- Shunyu Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Qiancheng Hu
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xinchuan Chen
- Department of Hematology, Sichuan University, Chengdu, Sichuan, China
| | - Nan Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Qiyue Huang
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Sirui Tan
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Minggang Su
- Department of Nuclear Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hongfeng Gou
- Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Reese M, Eichelmann AK, Nowacki TM, Pascher A, Sporn JC. The role of cytoreductive surgery and HIPEC for the treatment of primary and secondary peritoneal malignancies-experience from a tertiary care center in Germany. Langenbecks Arch Surg 2024; 409:113. [PMID: 38589714 DOI: 10.1007/s00423-024-03309-9] [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: 12/28/2023] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE Peritoneal surface malignancies (PSM) are commonly known to have a dismal prognosis. Over the past decades, novel techniques such as cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and pressurized intraperitoneal aerosol chemotherapy (PIPAC) have been introduced for the treatment of PSM which could improve the overall survival and quality of life of patients with PSM. The decision to proceed with CRS and HIPEC is often challenging due the complexity of the disease, the extent of the procedure, associated side effects, and potential risks. Here, we present our experience with CRS and HIPEC to add to the ongoing discussion about eligibility criteria, technical approach, and expected outcomes and contribute to the evolution of this powerful and promising tool in the multidisciplinary treatment of patients with primary and secondary PSM. METHODS A single-center retrospective chart review was conducted and included a total of 40 patients treated with CRS and HIPEC from April 2020 to September 2022 at the University Hospital Münster Department of Surgery. All patients had histologically confirmed primary or secondary peritoneal malignancies of various primary origins. RESULTS Our study included 22 patients with peritoneal metastases from gastric cancer (55%), 8 with pseudomyxoma peritonei (20%), 4 with mesothelioma of the peritoneum (10%), and 6 patients with PSM originating from other primary tumor locations. Median PCI at time of cytoreduction was 4 (0-25). Completeness of cytoreduction score was 0 in 37 patients (92.5%), 1 in two patients (5%), and 2 in one patient (2.5%). Median overall survival across all patients was 3.69 years. CONCLUSION Complete cytoreduction during CRS and HIPEC can be achieved for patients with low PCI, for patients with high PCI in low-grade malignancies, and even for patients with initially high PCI in high-grade malignancies following a significant reduction of cancer burden due to extensive preoperative treatment with PIPAC and systemic chemotherapy.
Collapse
Affiliation(s)
- Mikko Reese
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Ann-Kathrin Eichelmann
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Tobias M Nowacki
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, Albert-Schweitzer-Campus 1, Münster, 48149, Germany
- Department of Gastroenterology, UKM Marienhospital Steinfurt, Mauritiusstr. 5, Steinfurt, 48565, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany
| | - Judith C Sporn
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, Waldeyerstraße 1, 48149, Münster, Germany.
| |
Collapse
|
20
|
Liang Z, Wang P, Li Z, Wang D, Ma Q. Magnetoplasmonic Metasurface-Modulated Electrochemiluminescence Strategy for Extracellular Vesicle Detection. Anal Chem 2024; 96:4909-4917. [PMID: 38489746 DOI: 10.1021/acs.analchem.3c05650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Due to the ideal optical manipulation ability, the metasurface has broad prospects in the development of novel optical research. In particular, an active metasurface can control optical response through external stimulus, which has attracted great research interest. However, achieving effective modulation of the optical response is a significant challenge. In this work, we have developed a novel electrochemiluminescence (ECL) signal modulation strategy by an active magnetoplasmonic metasurface under an external magnetic field. The magnetoplasmonic metasurface was assembled based on yolk-shell Fe3O4@Au nanoparticles (Fe3O4@Au YS-NPs). On the one hand, the yolk-shell structure of Fe3O4@Au YS-NPs possessed the surface plasmon coupling effect and cavity-based Purcell effect, which provided high-intensity electromagnetic hot spots in the magnetoplasmonic metasurface. On the other hand, due to the strong magnetic response of the Fe3O4 core, the local magnetic field was induced by the external magnetic field, which further generated Lorentz force acting on the free electrons of Au nanoshells with strong optical anisotropy. The plasmon frequency of the metasurface can be effectively modulated by the Lorentz force effect. As a result, the ECL signal of nitrogen dots (N dots) was dynamically modulated and significantly enhanced at a specific polarization angle by the magnetoplasmonic metasurface under the variable external magnetic field. Based on the luminescence modulation ability and structure feature, the magnetoplasmonic metasurface was further established successfully as a sensing interface for gastric cancer (GC) extracellular vesicle (EV) detection. This study illustrated that the electromagnetic response of the active metasurface can effectively improve the optical modulation ability and luminescence sensing performance.
Collapse
Affiliation(s)
- Zihui Liang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Dongyu Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
21
|
Rijken A, Pape M, Simkens GA, de Hingh IHJT, Luyer MDP, van Sandick JW, van Laarhoven HWM, Verhoeven RHA, van Erning FN. Peritoneal metastases from gastric cancer in a nationwide cohort: Incidence, treatment and survival. Int J Cancer 2024; 154:992-1002. [PMID: 37916797 DOI: 10.1002/ijc.34780] [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: 04/22/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
The aims of this study were to investigate incidence, risk factors and treatment of synchronous or metachronous peritoneal metastases (PM) from gastric cancer and to estimate survival of these patients using population-based data. Patients diagnosed with gastric cancer in 2015 to 2016 were selected from the Netherlands Cancer Registry. The incidence of synchronous and metachronous PM were calculated. Multivariable regression analyses were performed to identify factors associated with the occurrence of PM. Treatment and survival were compared between patients with synchronous and metachronous PM. Of 2206 patients with gastric cancer, 741 (34%) were diagnosed with PM. Of these, 498 (23%) had synchronous PM. The cumulative incidence of metachronous PM in patients who underwent potentially curative treatment (n = 675) was 22.8% at 3 years. A factor associated with synchronous and metachronous PM was diffuse type histology. Patients diagnosed with synchronous PM more often received systemic treatment than patients with metachronous PM (35% vs 18%, respectively, P < .001). Median overall survival was comparable between synchronous and metachronous PM (3.2 vs 2.3 months, respectively, P = .731). Approximately one third of all patients with gastric cancer are diagnosed with PM, either at primary diagnosis or during 3-year follow-up after potentially curative treatment. Patients with metachronous PM less often received systemic treatment than those with synchronous PM but survival was comparable between both groups. Future trials are warranted to detect gastric cancer at an earlier stage and to examine strategies that lower the risk of peritoneal dissemination. Also, specific treatment options for patients with gastric PM should be further investigated.
Collapse
Affiliation(s)
- Anouk Rijken
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Marieke Pape
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- Amsterdam UMC location University of Amsterdam, Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Geert A Simkens
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- GROW-School for Oncology and Development Biology, Maastricht University, Maastricht, the Netherlands
| | - Misha D P Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Johanna W van Sandick
- Department of Surgery, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC location University of Amsterdam, Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Rob H A Verhoeven
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- Amsterdam UMC location University of Amsterdam, Medical Oncology, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Felice N van Erning
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| |
Collapse
|
22
|
Ghorbanzadeh S, Pourghasem N, Amiz R, Afsa M, Malekzadeh K. Investigating the Association between LncRNA NR2F2-AS1, miR-320b, and BMI1 in Gastric Cancer: Insights into Expression Profiles as Potential Biomarkers for Disease Management. Microrna 2024; 13:211-224. [PMID: 38952161 DOI: 10.2174/0122115366291818240606112725] [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: 03/02/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 07/03/2024]
Abstract
AIM This study aims to investigate the potential role of lncRNA NR2F2-AS1 in the development of gastric cancer by affecting the levels of miR-320b and BMI1. BACKGROUND Gastric cancer is a high-mortality malignancy, and understanding the underlying molecular mechanisms is crucial. Non-coding RNAs play an important role in gene expression, and their dysregulation can lead to tumor initiation and progression. OBJECTIVE This study aims to determine the pathological role of LncRNA NR2F2-AS1 in gastric cancer progression and its association with the clinicopathological characteristics of patients. METHODS Bioinformatics databases were used to predict the expression levels and interactions between the studied factors to achieve this objective. The expression pattern of NR2F2-AS1/miR- 320b/BMI1 in 40 pairs of tumor and adjacent normal tissues was examined using RT-PCR, IHC, and western blot. The correlation, ROC curve, and survival analyses were also conducted for the aforementioned factors. RESULTS The results showed an increase of more than 2-fold for BMI-1 and lncRNA NR2F2-AS1 in lower stages, and the elevation continued with the increasing stage of the disease. This correlated with significant downregulation of miR-320b and PTEN, indicating their association with gastric cancer progression and decreased patient survival. LncRNA NR2F2-AS1 acts as an oncogene by influencing the level of miR-320b, altering the amount of BMI1. A reduction in the amount of miR-320b against lncRNA NR2F2-AS1 and BMI1 directly correlates with a reduced overall survival rate of patients, especially if this disproportion is more than 3.0. ROC curve analysis indicated that alteration in the lncRNA NR2F2-AS1 level showed more than 98.0% sensitivity and specificity to differentiate the lower from higher stages of GC and predict the early onset of metastasis. CONCLUSION In conclusion, these results suggest that NR2F2-AS1/miR-320b/BMI1 has the potential to be a prognostic as well as diagnostic biomarker for gastric cancer.
Collapse
Affiliation(s)
- Shadi Ghorbanzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Molecular Diagnostic Department; Day Clinical Pathology Laboratory, Tabriz. Iran
| | - Navid Pourghasem
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Roghayeh Amiz
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Masoomeh Afsa
- Hormozgan Health Institute; Hormozgan University of Medical Sciences; Bandar Abbas; Iran
| | - Kianoosh Malekzadeh
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Hormozgan Health Institute; Hormozgan University of Medical Sciences; Bandar Abbas; Iran
| |
Collapse
|
23
|
Sarvestani AL, Gregory SN, Akmal SR, Hernandez JM, van der Sluis K, van Sandick JW. Gastrectomy + Cytoreductive Surgery + HIPEC for Gastric Cancer with Peritoneal Dissemination (PERISCOPE II). Ann Surg Oncol 2024; 31:28-30. [PMID: 37947975 DOI: 10.1245/s10434-023-14415-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/25/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Amber Leila Sarvestani
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie N Gregory
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sarfraz R Akmal
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan M Hernandez
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Karen van der Sluis
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Johanna W van Sandick
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Bencivenga M, Ministrini S, Morgagni P, Mura G, Marrelli D, Milandri C, Mazzei MA, Berselli M, Monti M, Graziosi L, Reddavid R, Rosa F, Solaini L, Donini A, Fumagalli Romario U, Roviello F, de Manzoni G, Tiberio GAM. Oligometastatic Gastric Cancer: Clinical Data from the Meta-Gastro Prospective Register of the Italian Research Group on Gastric Cancer. Cancers (Basel) 2023; 16:170. [PMID: 38201597 PMCID: PMC10778436 DOI: 10.3390/cancers16010170] [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: 11/18/2023] [Revised: 12/11/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Interest in the field of metastatic gastric cancer has grown in recent years, and the identification of oligometastatic patients plays a critical role as it consents to their inclusion in multimodal treatment strategies, which include systemic therapy but also surgery with curative intent. To collect sound clinical data on this subject, The Italian Research Group on Gastric Cancer developed a prospective multicentric observational register of metastatic gastric cancer patients called META-GASTRO. METHODS Data on 383 patients in Meta-Gastro were mined to help our understanding of oligometastatic, according to its double definition: quantitative/anatomical and dynamic. RESULTS the quantitative/anatomical definition applies to single-site metastases independently from the metastatic site (p < 0.001) to peritoneal metastases with PCI ≤ 12 (p = 0.009), to 1 or 2 hepatic metastases (p = 0.024) and nodal metastases in station 16 (p = 0.002). The dynamic definition applies to a percentage of cases variable according to the metastatic site: 8%, 13.5 and 23.8% for hepatic, lymphatic and peritoneal sites, respectively. In all cases, the OS of patients benefitting from conversion therapy was similar to those of cases deemed operable at diagnosis and operated after neoadjuvant chemotherapy. CONCLUSIONS META-GASTRO supports the two-fold definition of oligometastatic gastric cancer: the quantitative/anatomical one, which accounts for 30% of our population, and the dynamic one, observed in 16% of our cases.
Collapse
Affiliation(s)
- Maria Bencivenga
- General and Upper GI Surgery Division, Department of Surgery, University of Verona, 37125 Verona, Italy; (M.B.); (G.d.M.)
| | - Silvia Ministrini
- Surgical Unit, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, 25100 Brescia, Italy;
| | - Paolo Morgagni
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy; (P.M.); (L.S.)
| | - Gianni Mura
- Department of Surgery, San Donato Hospital, 52100 Arezzo, Italy;
| | - Daniele Marrelli
- Unit of General Surgery and Surgical Oncology, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (F.R.)
| | - Carlo Milandri
- Department of Oncology, San Donato Hospital, 52100 Arezzo, Italy;
| | - Maria Antonietta Mazzei
- Unit of Diagnostic Imaging, Department of Medical, Surgical and Neuro Sciences and of Radiological Sciences, Azienda Ospedaliero-Universitaria Senese, University of Siena, 53100 Siena, Italy;
| | - Mattia Berselli
- General Surgical Unit I, Department of Surgery, ASST Settelaghi-Varese, 21100 Varese, Italy;
| | - Manlio Monti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Luigina Graziosi
- Chirurgia Generale e d’Urgenza, Azienda Ospedaliera di Perugia, 06121 Perugia, Italy; (L.G.); (A.D.)
| | - Rossella Reddavid
- University of Turin, Department of Oncology, Division of Surgical Oncology and Digestive Surgery, San Luigi University Hospital, Orbassano, 10043 Torino, Italy;
| | - Fausto Rosa
- Department of Digestive Surgery, A. Gemelli Hospital, Catholic University, 00168 Roma, Italy;
| | - Leonardo Solaini
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, 47121 Forlì, Italy; (P.M.); (L.S.)
| | - Annibale Donini
- Chirurgia Generale e d’Urgenza, Azienda Ospedaliera di Perugia, 06121 Perugia, Italy; (L.G.); (A.D.)
| | | | - Franco Roviello
- Unit of General Surgery and Surgical Oncology, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (D.M.); (F.R.)
| | - Giovanni de Manzoni
- General and Upper GI Surgery Division, Department of Surgery, University of Verona, 37125 Verona, Italy; (M.B.); (G.d.M.)
| | - Guido Alberto Massimo Tiberio
- Surgical Unit, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili di Brescia, 25100 Brescia, Italy;
| |
Collapse
|
25
|
Guo J, Deng Z, Jin L, Yin S, Xiong Z, Wang C, Chen H, Luo D, Huang D, Peng J, Chen S, Lian L. Prognostic value of hyperthermic intraperitoneal chemotherapy in gastric cancer with synchronous peritoneal metastases: a real-world retrospective study. J Cancer Res Clin Oncol 2023; 149:17881-17896. [PMID: 37947869 DOI: 10.1007/s00432-023-05481-9] [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: 07/24/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
PURPOSE Peritoneal metastasis in gastric cancer (GC) is a late-stage condition with a poor prognosis. Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is a popular treatment for peritoneal metastases. Here, we aim to investigate the real-world application and efficacy of HIPEC alone for GC patients with synchronous peritoneal metastases. METHODS We conducted a retrospective analysis on GC patients with synchronous peritoneal metastasis at the Sixth Affiliated Hospital of Sun Yat-sen University between January 2011 and December 2022. Survival analyses and Cox regression models were performed based on overall survival (OS) and cancer-specific survival (CSS), and subgroup analysis was used to determine the prognostic value of HIPEC across different treatment. RESULTS We enrolled 250 patients, of whom 120 (48%) received HIPEC while 130 (52%) did not. HIPEC showed no survival benefit for GC patients (P = 0.220 for OS and P = 0.370 for CSS). However, subgroup analysis found that HIPEC can only improve OS and CSS when combined with primary tumor resection (P = 0.034 for OS and P = 0.036 for CSS). Moreover, survival analyses also demonstrated that HIPEC independently improved OS (HR for OS = 0.58, 95% CI 0.37-0.92, P = 0.020) and CSS (HR for CSS = 0.58, 95% CI 0.37-0.93, P = 0.024) for patients who underwent primary site resection, but not for those who did not. CONCLUSION HIPEC can improve survival in GC patients with synchronous peritoneal metastases who have primary tumor resection, but not in those without primary tumor resection.
Collapse
Affiliation(s)
- Jianping Guo
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zijian Deng
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Longyang Jin
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi Yin
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhizhong Xiong
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Caiqin Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huaxian Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dandong Luo
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dayin Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junsheng Peng
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Shi Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Lei Lian
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Rd., Guangzhou, Guangdong, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
26
|
Kryh-Jensen CG, Fristrup CW, Ainsworth AP, Detlefsen S, Mortensen MB, Pfeiffer P, Tarpgaard LS, Graversen M. What is long-term survival in patients with peritoneal metastasis from gastric, pancreatic, or colorectal cancer? A study of patients treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC). Pleura Peritoneum 2023; 8:147-155. [PMID: 38144215 PMCID: PMC10739291 DOI: 10.1515/pp-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Objectives A definition of long-term survival (LTS) in patients with peritoneal metastasis (PM) from gastric cancer (GC), pancreatic cancer (PC) or colorectal cancer (CRC) treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC) is lacking. We aimed to define LTS and investigate characteristics and treatment response in patients who reached LTS in data from two prospective trials. Methods Retrospective study of patients with GC-, PC-, or CRC-PM from the prospective PIPAC-OPC1 and PIPAC-OPC2 studies. The definition of LTS was based on published systematic reviews and randomized controlled trials. LTS was defined at the time point where 25 % of the patients were alive in these studies. Histology based response was evaluated by the mean Peritoneal Regression Grading Score (PRGS) using biopsies obtained prior to PIPAC 3, and defined by a mean PRGS of ≤2.0 or a decrease of mean PRGS of ≥1, compared to baseline. Results LTS was defined at 21 (GC), 15 (PC), and 24 (CRC) months. Fifty-one (47.2 %) patients (nine GC, 17 PC, 25 CRC) reached LTS calculated from the date of PM diagnosis. All but one received palliative chemotherapy before PIPAC, and 37 % received bidirectional treatment. More than 90 % of the LTS patients had response according to PRGS. The mOS from PIPAC 1 was 23.3, 12.4, and 28.5 months for GC, PC, and CRC LTS patients. Conclusions Patients with PM from GC, PC, and CRC treated with systemic chemotherapy and PIPAC can reach LTS and most show histological response. Causality must be further investigated.
Collapse
Affiliation(s)
- Charlotte G. Kryh-Jensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Claus W. Fristrup
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Alan P. Ainsworth
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Michael B. Mortensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Per Pfeiffer
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Line S. Tarpgaard
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- OPEN – Odense Patient Data Explorative Network, Odense University Hospital, Region of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
Winicki NM, Radomski SN, Florissi IS, Cloyd JM, Gutta G, Grotz TE, Scally CP, Fournier KF, Dineen SP, Powers BD, Veerapong J, Baumgartner JM, Clarke CN, Kothari AN, Maduekwe UN, Patel SH, Wilson GC, Schwartz P, Varley PR, Raoof M, Lee B, Malik I, Johnston FM, Greer JB. Neighborhood-Level Socioeconomic Disadvantage Predicts Outcomes in Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Peritoneal Malignancy. Ann Surg Oncol 2023; 30:7840-7847. [PMID: 37620532 PMCID: PMC10592201 DOI: 10.1245/s10434-023-14074-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) improves survival in select patients with peritoneal metastases (PM), but the impact of social determinants of health on CRS/HIPEC outcomes remains unclear. PATIENTS AND METHODS A retrospective review was conducted of a multi-institutional database of patients with PM who underwent CRS/HIPEC in the USA between 2000 and 2017. The area deprivation index (ADI) was linked to the patient's residential address. Patients were categorized as living in low (1-49) or high (50-100) ADI residences, with increasing scores indicating higher socioeconomic disadvantage. The primary outcome was overall survival (OS). Secondary outcomes included perioperative complications, hospital/intensive care unit (ICU) length of stay (LOS), and disease-free survival (DFS). RESULTS Among 1675 patients 1061 (63.3%) resided in low ADI areas and 614 (36.7%) high ADI areas. Appendiceal tumors (n = 1102, 65.8%) and colon cancer (n = 322, 19.2%) were the most common histologies. On multivariate analysis, high ADI was not associated with increased perioperative complications, hospital/ICU LOS, or DFS. High ADI was associated with worse OS (median not reached versus 49 months; 5 year OS 61.0% versus 28.2%, P < 0.0001). On multivariate Cox-regression analysis, high ADI (HR, 2.26; 95% CI 1.13-4.50; P < 0.001), cancer recurrence (HR, 2.26; 95% CI 1.61-3.20; P < 0.0001), increases in peritoneal carcinomatosis index (HR, 1.03; 95% CI 1.01-1.05; P < 0.001), and incomplete cytoreduction (HR, 4.48; 95% CI 3.01-6.53; P < 0.0001) were associated with worse OS. CONCLUSIONS Even after controlling for cancer-specific variables, adverse outcomes persisted in association with neighborhood-level socioeconomic disadvantage. The individual and structural-level factors leading to these cancer disparities warrant further investigation to improve outcomes for all patients with peritoneal malignancies.
Collapse
Affiliation(s)
- Nolan M Winicki
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shannon N Radomski
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isabella S Florissi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan M Cloyd
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Goutam Gutta
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | | | - Christopher P Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith F Fournier
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean P Dineen
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Benjamin D Powers
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jula Veerapong
- Department of Surgery, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Joel M Baumgartner
- Department of Surgery, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Callisia N Clarke
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anai N Kothari
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ugwuji N Maduekwe
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gregory C Wilson
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patrick Schwartz
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Patrick R Varley
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope, Duarte, CA, USA
| | - Byrne Lee
- Department of Surgery, Stanford University, Palo Alto, CA, USA
| | - Ibrahim Malik
- Department of Surgery, City of Hope, Duarte, CA, USA
| | - Fabian M Johnston
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan B Greer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Gastrointestinal Surgical Oncology, Peritoneal Surface Malignancy Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Ho SYA, Tay KV. Systematic review of diagnostic tools for peritoneal metastasis in gastric cancer-staging laparoscopy and its alternatives. World J Gastrointest Surg 2023; 15:2280-2293. [PMID: 37969710 PMCID: PMC10642463 DOI: 10.4240/wjgs.v15.i10.2280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Gastric cancer is one of the leading causes of cancer burden and mortality, often resulting in peritoneal metastasis in advanced stages with negative survival outcomes. Staging laparoscopy has become standard practice for suspected cases before a definitive gastrectomy or palliation. This systematic review aims to compare the efficacy of other diagnostic modalities instead of staging laparoscopy as the alternatives are able to reduce cost and invasive staging procedures. Recently, a radiomic model based on computed tomography and positron emission tomography (PET) has also emerged as another method to predict peritoneal metastasis. AIM To determine if the efficacy of computed tomography, magnetic resonance imaging and PET is comparable with staging laparoscopy. METHODS Articles comparing computed tomography, PET, magnetic resonance imaging, and radiomic models based on computed tomography and PET to staging laparoscopies were filtered out from the Cochrane Library, EMBASE, PubMed, Web of Science, and Reference Citations Analysis (https://www.referencecitationanalysis.com/). In the search for studies comparing computed tomography (CT) to staging laparoscopy, five retrospective studies and three prospective studies were found. Similarly, five retrospective studies and two prospective studies were also included for papers comparing CT to PET scans. Only one retrospective study and one prospective study were found to be suitable for papers comparing CT to magnetic resonance imaging scans. RESULTS Staging laparoscopy outperformed computed tomography in all measured aspects, namely sensitivity, specificity, positive predictive value and negative predictive value. Magnetic resonance imaging and PET produced mixed results, with the former shown to be only marginally better than computed tomography. CT performed slightly better than PET in most measured domains, except in specificity and true negative rates. We speculate that this may be due to the limited F-fluorodeoxyglucose uptake in small peritoneal metastases and in linitis plastica. Radiomic modelling, in its current state, shows promise as an alternative for predicting peritoneal metastases. With further research, deep learning and radiomic modelling can be refined and potentially applied as a preoperative diagnostic tool to reduce the need for invasive staging laparoscopy. CONCLUSION Staging laparoscopy was superior in all measured aspects. However, associated risks and costs must be considered. Refinements in radiomic modelling are necessary to establish it as a reliable screening technique.
Collapse
Affiliation(s)
| | - Kon Voi Tay
- Upper GI and Bariatric Division, General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
- Upper GI and Bariatric Division, General Surgery, Woodlands Health, Singapore 768024, Singapore
| |
Collapse
|
29
|
Luksta M, Bausys A, Bickaite K, Rackauskas R, Paskonis M, Luksaite-Lukste R, Ranceva A, Stulpinas R, Brasiuniene B, Baltruskeviciene E, Lachej N, Sabaliauskaite R, Bausys R, Tulyte S, Strupas K. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) with cisplatin and doxorubicin in combination with FOLFOX chemotherapy as a first-line treatment for gastric cancer patients with peritoneal metastases: single-arm phase II study. BMC Cancer 2023; 23:1032. [PMID: 37875869 PMCID: PMC10599063 DOI: 10.1186/s12885-023-11549-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/19/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) remains among the most common and most lethal cancers worldwide. Peritoneum is the most common site for distant dissemination. Standard treatment for GC peritoneal metastases (PM) is a systemic therapy, but treatment outcomes remain very poor, with median overall survival ranging between 3-9 months. Thus, novel treatment methods are necessary. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is the most novel technique for intraperitoneal chemotherapy. Some preliminary data suggest PIPAC can achieve improved long-term outcomes in patients with GC PM, especially when used in combination with systemic chemotherapy. However, there is a lack of data from well-design prospective studies that would confirm the efficacy of PIPAC and systemic therapy combination for first-line treatment. METHODS This study is an investigator-initiated single-arm, phase II trial to investigate the efficacy of PIPAC combined with systemic FOLFOX (5-fluorouracil, oxaliplatin, leucovorin) as a first-line treatment for GC PM. The study is conducted in 2 specialized GC treatment centers in Lithuania. It enrolls GC patients with histologically confirmed PM without prior treatment. The treatment protocol consists of PIPAC with cisplatin (10.5 mg/m2 body surface in 150 mL NaCl 0.9%) and doxorubicin (2.1 mg/m2 in 50 mL NaCl 0.9%) followed by 2 cycles of FOLFOX every 6-7 weeks. In total 3 PIPACs and 6 cycles of FOLFOX will be utilized. The primary outcome of the study is the objective response rate (ORR) according to RECIST v. 1.1 criteria (Eisenhauer et al., Eur J Cancer 45:228-47) in a CT scan performed 7 days after the 4th cycle of FOLFOX. Secondary outcomes include ORR after all experimental treatment, PIPAC characteristics, postoperative morbidity, histological and biochemical response, ascites volume, quality of life, overall survival, and toxicity. DISCUSSION This study aims to assess PIPAC and FOLFOX combination efficacy for previously untreated GC patients with PM. TRIAL REGISTRATION NCT05644249. Registered on December 9, 2022.
Collapse
Affiliation(s)
- Martynas Luksta
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101.
| | - Augustinas Bausys
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
- Centre for Visceral Medicine and Translational Research, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, Vilnius, 03101, Lithuania
| | - Klaudija Bickaite
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101
| | - Rokas Rackauskas
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101
| | - Marius Paskonis
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101
| | - Raminta Luksaite-Lukste
- Department of Radiology, Nuclear Medicine and Medical Physics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Anastasija Ranceva
- Hematology, Oncology, and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Rokas Stulpinas
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Birute Brasiuniene
- Department of Medical Oncology, National Cancer Institute, Vilnius, Lithuania
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Nadezda Lachej
- Department of Medical Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Rasa Sabaliauskaite
- Laboratory of Genetic Diagnostic, National Cancer Institute, Santariškių 1, Vilnius, LT-08406, Lithuania
| | - Rimantas Bausys
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Skaiste Tulyte
- Hematology, Oncology, and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Kestutis Strupas
- Clinic of Gastroenterology, Nephrourology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania, Ciurlionio str. 21, 03101
- Centre for Visceral Medicine and Translational Research, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, Vilnius, 03101, Lithuania
| |
Collapse
|
30
|
Carli M, Fini E, De Luca G, Scarselli M, Lamanna F, Vico T, Bocci G. Methadone dose escalation in patients with opioid use disorder and cancer as a strategy for controlling cancer-related pain: A case series. Palliat Support Care 2023:1-4. [PMID: 37786362 DOI: 10.1017/s1478951523001293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
OBJECTIVES Opioid use disorder (OUD) and cancer gained attention as co-occurring diseases in the last 2 decades due to the possible relationship between opioid prescriptions for cancer pain and the risk of developing substance use disorder in cancer patients. However, little is known about patients previously diagnosed with OUD who develop cancer and how to manage both OUD symptoms and control pain. METHODS The present case series deals with this subpopulation and proposes a dose escalation of methadone to control both the cancer-related pain and drug addiction symptoms. RESULTS This approach is peculiar because methadone is not used as a first-line treatment in cancer pain management and is not often used as a second-line treatment as well. Our 4 patients experienced good clinical control of symptoms and no major adverse reactions. SIGNIFICANCE OF RESULTS The subgroup of patients with OUD who develop cancer could be the perfect population to reconsider the use of methadone as a first-line treatment for cancer pain. Prospective studies are needed to evaluate the efficacy and safety of increasing doses of methadone in these patients to validate our clinical approach.
Collapse
Affiliation(s)
- Marco Carli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | - Elisabetta Fini
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Giulia De Luca
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Marco Scarselli
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Pisa, Italy
| | | | - Tiziana Vico
- Pisa Ser.D., Azienda USL Toscana Nord Ovest, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| |
Collapse
|
31
|
Choi W, Kim YH, Woo SM, Yu Y, Lee MR, Lee WJ, Chun JW, Sim SH, Chae H, Shim H, Lee KS, Kong SY. Establishment of Patient-Derived Organoids Using Ascitic or Pleural Fluid from Cancer Patients. Cancer Res Treat 2023; 55:1077-1086. [PMID: 37309112 PMCID: PMC10582554 DOI: 10.4143/crt.2022.1630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/07/2023] [Indexed: 06/14/2023] Open
Abstract
PURPOSE Patient-derived tumor cells can be a powerful resource for studying pathophysiological mechanisms and developing robust strategies for precision medicine. However, establishing organoids from patient-derived cells is challenging because of limited access to tissue specimens. Therefore, we aimed to establish organoids from malignant ascites and pleural effusions. MATERIALS AND METHODS Ascitic or pleural fluid from pancreatic, gastric, and breast cancer patients was collected and concentrated to culture tumor cells ex vivo. Organoids were considered to be successfully cultured when maintained for five or more passages. Immunohistochemical staining was performed to compare the molecular features, and drug sensitivity was assayed to analyze the clinical responses of original patients. RESULTS We collected 70 fluid samples from 58 patients (pancreatic cancer, n=39; gastric cancer, n=21; and breast cancer, n=10). The overall success rate was 40%; however, it differed with types of malignancy, with pancreatic, gastric, and breast cancers showing 48.7%, 33.3%, and 20%, respectively. Cytopathological results significantly differed between successful and failed cases (p=0.014). Immunohistochemical staining of breast cancer organoids showed molecular features identical to those of tumor tissues. In drug sensitivity assays, pancreatic cancer organoids recapitulated the clinical responses of the original patients. CONCLUSION Tumor organoids established from malignant ascites or pleural effusion of pancreatic, gastric, and breast cancers reflect the molecular characteristics and drug sensitivity profiles. Our organoid platform could be used as a testbed for patients with pleural and peritoneal metastases to guide precision oncology and drug discovery.
Collapse
Affiliation(s)
- Wonyoung Choi
- Center for Clinical Trials, National Cancer Center, Goyang, Korea
- Division of Cancer Biology, Cancer Molecular Biology Branch, Research Institute of National Cancer Center, Goyang, Korea
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
| | - Yun-Hee Kim
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
- Division of Technology Convergence, Cancer Molecular Imaging Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Sang Myung Woo
- Division of Rare and Refractory Cancer, Immuno-oncology Branch, Research Institute of National Cancer Center, Goyang, Korea
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer Center, Goyang, Korea
| | - Yebeen Yu
- Division of Rare and Refractory Cancer, Targeted Therapy Branch of Research Institute, National Cancer Center, Goyang, Korea
| | - Mi Rim Lee
- Division of Technology Convergence, Cancer Molecular Imaging Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer Center, Goyang, Korea
- Division of Clinical Research, Cancer Outcome & Quality Improvement Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer Center, Goyang, Korea
- Division of Clinical Research, Interventional Medicine Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Sung Hoon Sim
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
- Division of Rare and Refractory Cancer, Anticancer Resistance Branch, Research Institute of National Cancer Center, Goyang, Korea
| | - Heejung Chae
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
- Center for Cancer Data Center, National Cancer Control Institute of National Cancer Center, Goyang, Korea
| | - Hyoeun Shim
- Department of Laboratory Medicine, National Cancer Center, Goyang, Korea
| | - Keun Seok Lee
- Center for Clinical Trials, National Cancer Center, Goyang, Korea
- Center for Breast Cancer, National Cancer Center, Goyang, Korea
| | - Sun-Young Kong
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea
- Division of Rare and Refractory Cancer, Targeted Therapy Branch of Research Institute, National Cancer Center, Goyang, Korea
- Department of Laboratory Medicine, National Cancer Center, Goyang, Korea
| |
Collapse
|
32
|
Zhang Z, Yang A, Chaurasiya S, Park AK, Kim SI, Lu J, Valencia H, Fong Y, Woo Y. Anti-Tumor Immunogenicity of the Oncolytic Virus CF33-hNIS-antiPDL1 against Ex Vivo Peritoneal Cells from Gastric Cancer Patients. Int J Mol Sci 2023; 24:14189. [PMID: 37762490 PMCID: PMC10532045 DOI: 10.3390/ijms241814189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
We studied the immunotherapeutic potential of CF33-hNIS-antiPDL1 oncolytic virus (OV) against gastric cancer with peritoneal metastasis (GCPM). We collected fresh malignant ascites (MA) or peritoneal washings (PW) during routine paracenteses and diagnostic laparoscopies from GC patients (n = 27). Cells were analyzed for cancer cell markers and T cells, or treated with PBS, CF33-GFP, or CF33-hNIS-antiPDL1 (MOI = 3). We analyzed infectivity, replication, cytotoxicity, CD107α upregulation of CD8+ and CD4+ T cells, CD274 (PD-L1) blockade of cancer cells by virus-encoded anti-PD-L1 scFv, and the release of growth factors and cytokines. We observed higher CD45-/large-size cells and lower CD8+ T cell percentages in MA than PW. CD45-/large-size cells were morphologically malignant and expressed CD274 (PD-L1), CD252 (OX40L), and EGFR. CD4+ and CD8+ T cells did not express cell surface exhaustion markers. Virus infection and replication increased cancer cell death at 15 h and 48 h. CF33-hNIS-antiPDL1 treatment produced functional anti-PD-L1 scFv, which blocked surface PD-L1 binding of live cancer cells and increased CD8+CD107α+ and CD4+CD107α+ T cell percentages while decreasing EGF, PDGF, soluble anti-PD-L1, and IL-10. CF33-OVs infect, replicate in, express functional proteins, and kill ex vivo GCPM cells with immune-activating effects. CF33-hNIS-antiPDL1 displays real potential for intraperitoneal GCPM therapy.
Collapse
Affiliation(s)
- Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Anthony K. Park
- Cancer Immunotherapeutics Program, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Hannah Valencia
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (Z.Z.); (A.Y.); (S.C.); (S.-I.K.); (J.L.); (H.V.); (Y.F.)
- Cancer Immunotherapeutics Program, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA 91010, USA;
| |
Collapse
|
33
|
Casella F, Bencivenga M, Brancato G, Torroni L, Ridolfi C, Puccio C, Alloggio M, Meloni F, Fusario D, Marrelli D, Giacopuzzi S, Roviello F, de Manzoni G. Bidirectional Approach with PIPAC and Systemic Chemotherapy for Patients with Synchronous Gastric Cancer Peritoneal Metastases (GCPM). Ann Surg Oncol 2023; 30:5733-5742. [PMID: 37270440 PMCID: PMC10409663 DOI: 10.1245/s10434-023-13572-7] [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: 03/09/2023] [Accepted: 04/12/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND This study evaluated the efficacy of pressurized intraperitoneal aerosol chemotherapy (PIPAC) with systemic chemotherapy as a bidirectional approach for gastric cancer (GC) patients with synchronous peritoneal metastases (SPM). METHODS A retrospective analysis of a prospective PIPAC database was queried for patients who underwent a bidirectional approach between October 2019 and April 2022 at two high-volume GC surgery units in Italy (Verona and Siena). Surgical and oncological outcomes were analyzed. RESULTS Between October 2019 and April 2022, 74 PIPAC procedures in 42 consecutive patients with Eastern Cooperative Oncology Group performance status ≤2 were performed-32 patients treated in Verona and 10 in Siena. Twenty-seven patients (64%) were female and median age at first PIPAC was 60.5 years (I-III quartiles: 49-68 years). Median Peritoneal Cancer Index (PCI) was 16 (I-III quartiles: 8-26) and 25 patients (59%) had at least two PIPAC procedures. Major complications according to the Common Terminology Criteria for Adverse Events (CTCAE; 3 and 4) occurred in three (4%) procedures, and, according to the Clavien-Dindo classification (>3a), one (1%) severe complication occurred. There were no reoperations or deaths within 30 days. Median overall survival (mOS) from diagnosis was 19.6 months (range 14-24), and mOS from first PIPAC was 10.5 months (range 7-13). Excluding cases with very heavy metastatic peritoneal burden, with PCI from 2 to 26, treated with more than one PIPAC, mOS from diagnosis was 22 months (range 14-39). Eleven patients (26%) underwent curative-intent surgery after a bidirectional approach. R0 was achieved in nine (82%) patients and complete pathological response was obtained in three (27%) cases. CONCLUSIONS Patient selection is associated with bidirectional approach efficacy and feasibility for SPM GC treatment, which may allow potentially curative surgical radicalization in highly selected cases.
Collapse
Affiliation(s)
| | - Maria Bencivenga
- Upper G.I. Surgery Division, University of Verona, Verona, Italy.
| | - Giorgio Brancato
- Upper G.I. Surgery Division, University of Verona, Verona, Italy
| | - Lorena Torroni
- Department of Diagnostics and Public Health, Unit of Epidemiology and Medical Statistics, University of Verona, Verona, Italy
| | - Cecilia Ridolfi
- Upper G.I. Surgery Division, University of Verona, Verona, Italy
| | - Carmelo Puccio
- Upper G.I. Surgery Division, University of Verona, Verona, Italy
| | | | - Francesca Meloni
- Upper G.I. Surgery Division, University of Verona, Verona, Italy
| | - Daniele Fusario
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Siena, Italy
| | - Daniele Marrelli
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Siena, Italy
| | | | - Franco Roviello
- Department of Medicine, Surgery and Neurosciences, Unit of General Surgery and Surgical Oncology, University of Siena, Siena, Italy
| | | |
Collapse
|
34
|
Díaz del Arco C, Ortega Medina L, Estrada Muñoz L, Molina Roldán E, García Gómez de las Heras S, Fernández Aceñero MJ. Impact of Age at Diagnosis on Clinicopathological Features, Prognosis, and Management of Gastric Cancer: A Retrospective Single-Center Experience from Spain. Cancers (Basel) 2023; 15:4241. [PMID: 37686517 PMCID: PMC10486869 DOI: 10.3390/cancers15174241] [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: 07/19/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The impact of age on various aspects of gastric cancer (GC) remains controversial. Clarifying this issue can improve our understanding of the disease, refine risk stratification models, and aid in personalized therapeutic approaches. This study aimed to evaluate the influence of age at diagnosis on the clinicopathological features, prognosis, and management of a specific cohort of Spanish patients with resected GC. The study encompassed 315 patients treated at a single tertiary hospital in Spain, divided into two age-based subgroups: ≤65 years and >65 years. The mean and median ages at diagnosis were 72 and 76 years. Most tumors were diagnosed at pT3 stage (49.2%), and 59.6% of patients had lymph node metastases. 21.3% of cases were diagnosed with GC at age ≤ 65 years. Younger patients showed a significantly higher prevalence of flat, diffuse, high-grade tumors, signet-ring cells, perineural infiltration, D2 lymphadenectomies, and adjuvant therapy. They also exhibited a higher rate of recurrences, but had a significantly longer follow-up. Kaplan-Meier curves indicated no significant prognostic differences based on age. Finally, age did not independently predict overall survival or disease-free survival. Our results suggest that younger patients may require more aggressive treatment due to adverse clinicopathologic features, but the lack of prognostic differences among age groups in our cohort indicates the need for further investigation into the complex interplay between age, clinicopathologic factors, and long-term outcomes in GC.
Collapse
Affiliation(s)
- Cristina Díaz del Arco
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (L.O.M.); (M.J.F.A.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Luis Ortega Medina
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (L.O.M.); (M.J.F.A.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
| | - Lourdes Estrada Muñoz
- Department of Basic Medical Sciences, School of Medicine, Rey Juan Carlos University, Móstoles, 28933 Madrid, Spain; (L.E.M.); (S.G.G.d.l.H.)
- Department of Pathology, Rey Juan Carlos Hospital, Móstoles, 28933 Madrid, Spain
| | - Elena Molina Roldán
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
- Biobank, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Soledad García Gómez de las Heras
- Department of Basic Medical Sciences, School of Medicine, Rey Juan Carlos University, Móstoles, 28933 Madrid, Spain; (L.E.M.); (S.G.G.d.l.H.)
| | - María Jesús Fernández Aceñero
- Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (L.O.M.); (M.J.F.A.)
- Department of Pathology, Hospital Clínico San Carlos, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain;
| |
Collapse
|
35
|
Ramos MFKP, Pereira MA, Charruf AZ, Victor CR, Gregorio JVAM, Alban LBV, Moniz CMV, Zilberstein B, Mello ESD, Hoff PMG, Ribeiro Junior U, Dias AR. INTRAPERITONEAL CHEMOTHERAPY FOR GASTRIC CANCER WITH PERITONEAL CARCINOMATOSIS: STUDY PROTOCOL OF A PHASE II TRIAL. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2023; 36:e1744. [PMID: 37466566 PMCID: PMC10356002 DOI: 10.1590/0102-672020230026e1744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/26/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Peritoneal carcinomatosis in gastric cancer is considered a fatal disease, without expectation of definitive cure. As systemic chemotherapy is not sufficient to contain the disease, a multimodal approach associating intraperitoneal chemotherapy with surgery may represent an alternative for these cases. AIMS The aim of this study was to investigate the role of intraperitoneal chemotherapy in stage IV gastric cancer patients with peritoneal metastasis. METHODS This study is a single institutional single-arm prospective clinical trial phase II (NCT05541146). Patients with the following inclusion criteria undergo implantation of a peritoneal catheter for intraperitoneal chemotherapy: Stage IV gastric adenocarcinoma; age 18-75 years; Peritoneal carcinomatosis with peritoneal cancer index<12; Eastern Cooperative Oncology Group 0/1; good clinical status; and lab exams within normal limits. The study protocol consists of four cycles of intraperitoneal chemotherapy with paclitaxel associated with systemic chemotherapy. After treatment, patients with peritoneal response assessed by staging laparoscopy undergo conversion gastrectomy. RESULTS The primary outcome is the rate of complete peritoneal response. Progression-free and overall survivals are other outcomes evaluated. The study started in July 2022, and patients will be screened for inclusion until 30 are enrolled. CONCLUSIONS Therapies for advanced gastric cancer patients have been evaluated in clinical trials but without success in patients with peritoneal metastasis. The treatment proposed in this trial can be promising, with easy catheter implantation and ambulatory intraperitoneal chemotherapy regime. Verifying the efficacy and safety of paclitaxel with systemic chemotherapy is an important progress that this study intends to investigate.
Collapse
Affiliation(s)
| | - Marina Alessandra Pereira
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Gastroenterology - São Paulo (SP), Brazil
| | - Amir Zeide Charruf
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Gastroenterology - São Paulo (SP), Brazil
| | - Carolina Ribeiro Victor
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Radiology and Oncology - São Paulo (SP), Brazil
| | | | - Luciana Bastos Valente Alban
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Radiology and Oncology - São Paulo (SP), Brazil
| | - Camila Motta Venchiarutti Moniz
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Radiology and Oncology - São Paulo (SP), Brazil
| | - Bruno Zilberstein
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Gastroenterology - São Paulo (SP), Brazil
| | - Evandro Sobroza de Mello
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Pathology - São Paulo (SP), Brazil
| | - Paulo Marcelo Gehm Hoff
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Radiology and Oncology - São Paulo (SP), Brazil
| | - Ulysses Ribeiro Junior
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Gastroenterology - São Paulo (SP), Brazil
| | - Andre Roncon Dias
- Universidade de São Paulo, Cancer Institute, Faculty of Medicine, Department of Gastroenterology - São Paulo (SP), Brazil
| |
Collapse
|
36
|
Zhang F, Wu G, Chen N, Li R. The predictive value of radiomics-based machine learning for peritoneal metastasis in gastric cancer patients: a systematic review and meta-analysis. Front Oncol 2023; 13:1196053. [PMID: 37465109 PMCID: PMC10352083 DOI: 10.3389/fonc.2023.1196053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023] Open
Abstract
Background For patients with gastric cancer (GC), effective preoperative identification of peritoneal metastasis (PM) remains a severe challenge in clinical practice. Regrettably, effective early identification tools are still lacking up to now. With the popularization and application of radiomics method in tumor management, some researchers try to introduce it into the early identification of PM in patients with GC. However, due to the complexity of radiomics, the value of radiomics method in the early identification of PM in GC patients remains controversial. Therefore, this systematic review was conducted to explore the feasibility of radiomics in the early identification of PM in GC patients. Methods PubMed, Cochrane, Embase and the Web of Science were comprehensively and systematically searched up to 25 July, 2022 (CRD42022350512). The quality of the included studies was assessed using the radiomics quality score (RQS). To discuss the superiority in diagnostic accuracy of radiomics-based machine learning, a subgroup analysis was performed by machine learning (ML) based on clinical features, radiomics features, and radiomics + clinical features. Results Finally, 11 eligible original studies covering 78 models were included in this systematic review. According to the meta-analysis, the radiomics + clinical features model had a c-index of 0.919 (95% CI: 0.871-0.969), pooled sensitivity and specificity of 0.90 (0.83-0.94) and 0.87 (0.78-0.92), respectively, in the training set, and a c- index of 0.910 (95% CI: 0.886-0.934), pooled sensitivity and specificity of 0.78 (0.71-0.84) and 0.83 (0.74-0.89), respectively, in the validation set. Conclusions The ML methods based on radiomics + clinical features had satisfactory accuracy for the early diagnosis of PM in GC patients, and can be used as an auxiliary diagnostic tool for clinicians. However, the lack of guidelines for the proper operation of radiomics has led to the diversification of radiomics methods, which seems to limit the development of radiomics. Even so, the clinical application value of radiomics cannot be ignored. The standardization of radiomics research is required in the future for the wider application of radiomics by developing intelligent tools of radiomics. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=350512, identifier CRD42022350512.
Collapse
|
37
|
Yang A, Zhang Z, Chaurasiya S, Park AK, Jung A, Lu J, Kim SI, Priceman S, Fong Y, Woo Y. Development of the oncolytic virus, CF33, and its derivatives for peritoneal-directed treatment of gastric cancer peritoneal metastases. J Immunother Cancer 2023; 11:e006280. [PMID: 37019471 PMCID: PMC10083877 DOI: 10.1136/jitc-2022-006280] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) that metastasizes to the peritoneum is fatal. CF33 and its genetically modified derivatives show cancer selectivity and oncolytic potency against various solid tumors. CF33-hNIS and CF33-hNIS-antiPDL1 have entered phase I trials for intratumoral and intravenous treatments of unresectable solid tumors (NCT05346484) and triple-negative breast cancer (NCT05081492). Here, we investigated the antitumor activity of CF33-oncolytic viruses (OVs) against GC and CF33-hNIS-antiPDL1 in the intraperitoneal (IP) treatment of GC peritoneal metastases (GCPM). METHODS We infected six human GC cell lines AGS, MKN-45, MKN-74, KATO III, SNU-1, and SNU-16 with CF33, CF33-GFP, or CF33-hNIS-antiPDL1 at various multiplicities of infection (0.01, 0.1, 1.0, and 10.0), and performed viral proliferation and cytotoxicity assays. We used immunofluorescence imaging and flow cytometric analysis to verify virus-encoded gene expression. We evaluated the antitumor activity of CF33-hNIS-antiPDL1 following IP treatment (3×105 pfu × 3 doses) in an SNU-16 human tumor xenograft model using non-invasive bioluminescence imaging. RESULTS CF33-OVs showed dose-dependent infection, replication, and killing of both diffuse and intestinal subtypes of human GC cell lines. Immunofluorescence imaging showed virus-encoded GFP, hNIS, and anti-PD-L1 antibody scFv expression in CF33-OV-infected GC cells. We confirmed GC cell surface PD-L1 blockade by virus-encoded anti-PD-L1 scFv using flow cytometry. In the xenograft model, CF33-hNIS-antiPDL1 (IP; 3×105 pfu × 3 doses) treatment significantly reduced peritoneal tumors (p<0.0001), decreased amount of ascites (62.5% PBS vs 25% CF33-hNIS-antiPDL1) and prolonged animal survival. At day 91, seven out of eight mice were alive in the virus-treated group versus one out of eight in the control group (p<0.01). CONCLUSIONS Our results show that CF33-OVs can deliver functional proteins and demonstrate effective antitumor activity in GCPM models when delivered intraperitoneally. These preclinical results will inform the design of future peritoneal-directed therapy in GCPM patients.
Collapse
Affiliation(s)
- Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Anthony K Park
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Audrey Jung
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Saul Priceman
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
38
|
Guchelaar NAD, Noordman BJ, Koolen SLW, Mostert B, Madsen EVE, Burger JWA, Brandt-Kerkhof ARM, Creemers GJ, de Hingh IHJT, Luyer M, Bins S, van Meerten E, Lagarde SM, Verhoef C, Wijnhoven BPL, Mathijssen RHJ. Intraperitoneal Chemotherapy for Unresectable Peritoneal Surface Malignancies. Drugs 2023; 83:159-180. [PMID: 36633826 PMCID: PMC9908703 DOI: 10.1007/s40265-022-01828-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Malignancies of the peritoneal cavity are associated with a dismal prognosis. Systemic chemotherapy is the gold standard for patients with unresectable peritoneal disease, but its intraperitoneal effect is hampered by the peritoneal-plasma barrier. Intraperitoneal chemotherapy, which is administered repeatedly into the peritoneal cavity through a peritoneal implanted port, could provide a novel treatment modality for this patient population. This review provides a systematic overview of intraperitoneal used drugs, the performed clinical studies so far, and the complications of the peritoneal implemental ports. Several anticancer drugs have been studied for intraperitoneal application, with the taxanes paclitaxel and docetaxel as the most commonly used drug. Repeated intraperitoneal chemotherapy, mostly in combination with systemic chemotherapy, has shown promising results in Phase I and Phase II studies for several tumor types, such as gastric cancer, ovarian cancer, colorectal cancer, and pancreatic cancer. Two Phase III studies for intraperitoneal chemotherapy in gastric cancer have been performed so far, but the results regarding the superiority over standard systemic chemotherapy alone, are contradictory. Pressurized intraperitoneal administration, known as PIPAC, is an alternative way of administering intraperitoneal chemotherapy, and the first prospective studies have shown a tolerable safety profile. Although intraperitoneal chemotherapy might be a standard treatment option for patients with unresectable peritoneal disease, more Phase II and Phase III studies focusing on tolerability profiles, survival rates, and quality of life are warranted in order to establish optimal treatment schedules and to establish a potential role for intraperitoneal chemotherapy in the approach to unresectable peritoneal disease.
Collapse
Affiliation(s)
- Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - Bo J Noordman
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.,Department of Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Eva V E Madsen
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jacobus W A Burger
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Alexandra R M Brandt-Kerkhof
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands.,Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Misha Luyer
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Sjoerd M Lagarde
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Bas P L Wijnhoven
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Tissue factor-induced fibrinogenesis mediates cancer cell clustering and multiclonal peritoneal metastasis. Cancer Lett 2023; 553:215983. [PMID: 36404569 DOI: 10.1016/j.canlet.2022.215983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 11/02/2022]
Abstract
Peritoneal metastasis is one of the most frequent causes of death in several types of advanced cancers; however, the underlying molecular mechanisms remain largely unknown. In this study, we exploited multicolor fluorescent lineage tracking to investigate the clonality of peritoneal metastasis in mouse xenograft models. When peritoneal metastasis was induced by intraperitoneal or orthotopic injection of multicolored cancer cells, each peritoneally metastasized tumor displayed multicolor fluorescence regardless of metastasis sites, indicating that it consists of multiclonal cancer cell populations. Multicolored cancer cell clusters form within the peritoneal cavity and collectively attach to the peritoneum. In vitro, peritoneal lavage fluid or cleared ascitic fluid derived from cancer patients induces cancer cell clustering, which is inhibited by anticoagulants. Cancer cell clusters formed in vitro and in vivo are associated with fibrin formation. Furthermore, tissue factor knockout in cancer cells abrogates cell clustering, peritoneal attachment, and peritoneal metastasis. Thus, we propose that cancer cells activate the coagulation cascade via tissue factor to form fibrin-mediated cell clusters and promote peritoneal attachment; these factors lead to the development of multiclonal peritoneal metastasis and may be therapeutic targets.
Collapse
|
40
|
Tabari A, Chan SM, Omar OMF, Iqbal SI, Gee MS, Daye D. Role of Machine Learning in Precision Oncology: Applications in Gastrointestinal Cancers. Cancers (Basel) 2022; 15:cancers15010063. [PMID: 36612061 PMCID: PMC9817513 DOI: 10.3390/cancers15010063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) cancers, consisting of a wide spectrum of pathologies, have become a prominent health issue globally. Despite medical imaging playing a crucial role in the clinical workflow of cancers, standard evaluation of different imaging modalities may provide limited information. Accurate tumor detection, characterization, and monitoring remain a challenge. Progress in quantitative imaging analysis techniques resulted in "radiomics", a promising methodical tool that helps to personalize diagnosis and treatment optimization. Radiomics, a sub-field of computer vision analysis, is a bourgeoning area of interest, especially in this era of precision medicine. In the field of oncology, radiomics has been described as a tool to aid in the diagnosis, classification, and categorization of malignancies and to predict outcomes using various endpoints. In addition, machine learning is a technique for analyzing and predicting by learning from sample data, finding patterns in it, and applying it to new data. Machine learning has been increasingly applied in this field, where it is being studied in image diagnosis. This review assesses the current landscape of radiomics and methodological processes in GI cancers (including gastric, colorectal, liver, pancreatic, neuroendocrine, GI stromal, and rectal cancers). We explain in a stepwise fashion the process from data acquisition and curation to segmentation and feature extraction. Furthermore, the applications of radiomics for diagnosis, staging, assessment of tumor prognosis and treatment response according to different GI cancer types are explored. Finally, we discussed the existing challenges and limitations of radiomics in abdominal cancers and investigate future opportunities.
Collapse
Affiliation(s)
- Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| | - Shin Mei Chan
- Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06510, USA
| | - Omar Mustafa Fathy Omar
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Shams I. Iqbal
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Michael S. Gee
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Mommersteeg MC, Kies DA, van der Laan J, Wonders J. Linitis plastica of the rectum secondary to prostate carcinoma. BMJ Case Rep 2022; 15:e248462. [PMID: 36460309 PMCID: PMC9723822 DOI: 10.1136/bcr-2021-248462] [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] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
Linitis plastica is an intramural carcinoma that may occur in any hollow organ. Rectal linitis plastica (RLP) is a morphological variant cancer that may occur as a primary form of cancer or secondary as a metastasis of a primary malignancy. We report the case of a man in his 70s with RLP secondary to prostate carcinoma who was initially suspected to have an obstructing rectal adenocarcinoma. During colonoscopy a segment of cobblestone mucosa was seen in the distal rectum. Subsequent imaging showed enhancement of all wall-layers of the rectum and diffuse retroperitoneal fat infiltration with traction on both ureters. A prostate-specific membrane antigen scan confirmed RLP secondary to a prostate carcinoma mimicking the clinical and radiological signs of an obstructing rectal carcinoma with retroperitoneal fibrosis.This case emphasises the possible pitfalls in the diagnosis of RLP and the importance of advanced imaging techniques, such as MRI, as well as appropriate histological samples. The patient underwent androgen deprivation therapy to which RLP responded well and neither systemic chemotherapy or surgery was necessary.
Collapse
Affiliation(s)
- Michiel C Mommersteeg
- Department of Gastroenterology and Hepatology, HagaZiekenhuis, Den Haag, Zuid-Holland, The Netherlands
| | - Dennis A Kies
- Department of Radiology, HagaZiekenhuis, Den Haag, Zuid-Holland, The Netherlands
| | - Jaap van der Laan
- Department of Pathology, HagaZiekenhuis Locatie Leyweg, Den Haag, Zuid-Holland, The Netherlands
| | - Janneke Wonders
- Department of Gastroenterology and Hepatology, HagaZiekenhuis, Den Haag, Zuid-Holland, The Netherlands
| |
Collapse
|
42
|
Case A, Prosser S, Peters CJ, Adams R, Gwynne S. Pressurised intraperitoneal aerosolised chemotherapy (PIPAC) for gastric cancer with peritoneal metastases: A systematic review by the PIPAC UK collaborative. Crit Rev Oncol Hematol 2022; 180:103846. [PMID: 36257535 DOI: 10.1016/j.critrevonc.2022.103846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/30/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Gastric cancer with peritoneal metastases (GCPM) carries a poor prognosis. Pressurised Intraperitoneal Aerosolised Chemotherapy (PIPAC) offers pharmacokinetic advantages over intravenous therapy, resulting in higher chemotherapy concentrations in peritoneal deposits, and potentially reduced systemic absorption/toxicity. This review evaluates efficacy, tolerability and impact on quality of life (QOL) of PIPAC for GCPM. METHODS Following registration with PROSPERO (CRD42021281500), MEDLINE, EMBASE and The Cochrane Library were searched for PIPAC in patients with peritoneal metastases, in accordance with PRISMA standards RESULTS: Across 18 included reports representing 751 patients with GCPM (4 prospective, 11 retrospective, 3 abstracts, no phase III studies), median overall survival (mOS) was 8 - 19.1 months, 1-year OS 49.8-77.9%, complete response (PRGS1) 0-35% and partial response (PRGS2/3) 0-83.3%. Grade 3 and 4 toxicity was 0.7-25% and 0-4.1% respectively. Three studies assessing QOL reported no significant difference. CONCLUSION PIPAC may offer promising survival benefits, toxicity, and QOL for GCPM.
Collapse
Affiliation(s)
- A Case
- South West Wales Cancer Centre, Singleton Hospital, Sketty Lane, Swansea SA2 8QA, UK; Swansea University Medical School, Grove Building, Singleton Park, SA2 8PP, UK.
| | - S Prosser
- South West Wales Cancer Centre, Singleton Hospital, Sketty Lane, Swansea SA2 8QA, UK
| | - C J Peters
- Department of Surgery and Cancer, Imperial College London, St Marys Hospital, Praed Street, London W2 1NY, UK
| | - R Adams
- Centre for Trials Research, Cardiff University and Velindre Cancer Centre, Velindre Road, Whitchurch CF14 2TL, UK
| | - S Gwynne
- South West Wales Cancer Centre, Singleton Hospital, Sketty Lane, Swansea SA2 8QA, UK; Swansea University Medical School, Grove Building, Singleton Park, SA2 8PP, UK
| |
Collapse
|
43
|
Zhou H, Gao Z, Sun A, Huang H, Zhang X, Li K. Editorial: Peritoneal Metastasis of Gastric Cancer: From Basic Research to Clinical Application. Front Oncol 2022; 12:880497. [PMID: 35837111 PMCID: PMC9274279 DOI: 10.3389/fonc.2022.880497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Affiliation(s)
- Heng Zhou
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Ziming Gao
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Anqi Sun
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Haibo Huang
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
- *Correspondence: Kai Li, ; Xiaotian Zhang,
| | - Kai Li
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Kai Li, ; Xiaotian Zhang,
| |
Collapse
|
44
|
Wintjens AGWE, Simkens GA, Fransen PPKH, Serafras N, Lenaerts K, Franssen GHLM, de Hingh IHJT, Dankers PYW, Bouvy ND, Peeters A. Intraperitoneal drug delivery systems releasing cytostatic agents to target gastro-intestinal peritoneal metastases in laboratory animals: a systematic review. Clin Exp Metastasis 2022; 39:541-579. [PMID: 35737252 PMCID: PMC9338897 DOI: 10.1007/s10585-022-10173-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022]
Abstract
For peritoneal metastases (PM), there are few curative treatment options, and they are only available for a select patient group. Recently, new therapies have been developed to deliver intraperitoneal chemotherapy for a prolonged period, suitable for a larger patient group. These drug delivery systems (DDSs) seem promising in the experimental setting. Many types of DDSs have been explored in a variety of animal models, using different cytostatics. This review aimed to provide an overview of animal studies using DDSs containing cytostatics for the treatment of gastro-intestinal PM and identify the most promising therapeutic combinations. The review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) guidelines. The 35 studies included revealed similar results: using a cytostatic-loaded DDS to treat PM resulted in a higher median survival time (MST) and a lower intraperitoneal tumor load compared to no treatment or treatment with a ‘free’ cytostatic or an unloaded DDS. In 65% of the studies, the MST was significantly longer and in 24% the tumor load was significantly lower in the animals treated with cytostatic-loaded DDS. The large variety of experimental setups made it impossible to identify the most promising DDS-cytostatic combination. In most studies, the risk of bias was unclear due to poor reporting. Future studies should focus more on improving the clinical relevance of the experiments, standardizing the experimental study setup, and improving their methodological quality and reporting.
Collapse
Affiliation(s)
- Anne G W E Wintjens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands. .,Department of Surgery, Maastricht University Medical Centre, PO Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Geert A Simkens
- Department of Surgery, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
| | | | - Narcis Serafras
- Department of Surgery, Maastricht University Medical Centre, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Kaatje Lenaerts
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,Department of Surgery, Maastricht University Medical Centre, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Gregor H L M Franssen
- Department of Education, Content & Support, University Library, Maastricht University, Maastricht, The Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital Eindhoven, Eindhoven, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Nicole D Bouvy
- Department of Surgery, Maastricht University Medical Centre, PO Box 616, 6200 MD, Maastricht, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Andrea Peeters
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
45
|
Casella F, Bencivenga M, Rosati R, Fumagalli UR, Marrelli D, Pacelli F, Macrì A, Donini A, Torroni L, Pavarana M, De Manzoni G. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) in multimodal therapy for patients with oligometastatic peritoneal gastric cancer: a randomized multicenter phase III trial PIPAC VEROne. Pleura Peritoneum 2022; 7:135-141. [PMID: 36159218 PMCID: PMC9467896 DOI: 10.1515/pp-2022-0111] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/03/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objectives
Peritoneal carcinomatosis is the most frequent site of metastases in patients with gastric cancer. Current standard treatment is palliative systemic chemotherapy with very poor prognosis. Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) resulted in long-term benefits in selected patients. Among patients with peritoneal carcinomatosis, a distinctive subset is oligometastatic disease which is characterized by low metastatic burden. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a recent technique of intraperitoneal chemotherapy used in combination with systemic chemotherapy with promising results.
Methods
PIPAC VER-One is a prospective, randomized, multicenter phase III clinical trial that aims to evaluate the effectiveness of the use of PIPAC in combination with systemic chemotherapy in patients with gastric cancer and synchronous positive peritoneal cytology and/or limited peritoneal metastases (peritoneal cancer index [PCI] ≤6). Patients will be randomized into two arms: arm A (control) treated with standard systemic chemotherapy and arm B (experimental) treated with a bidirectional scheme including PIPAC and systemic chemotherapy.
Results
Primary endpoint is the secondary resectability rate. Secondary endpoints are: overall survival (OS), pregression-free survival (PFS), disease-free survival (DFS), histological response assessed both on primary tumor and peritoneal lesions, quality of life (QoL), complication rate (CTCAE v5), and incremental cost-effectiveness ratios (ICER).
Conclusions
The role of PIPAC in multimodal treatment for oligometastatic gastric cancer will be investigated in this trial.
Collapse
Affiliation(s)
- Francesco Casella
- Department and of Surgical, General and Upper GI Surgery Division, Odontostomatologic, Maternal and Child Sciences , University of Verona , Verona , Italy
| | - Maria Bencivenga
- Department and of Surgical, General and Upper GI Surgery Division, Odontostomatologic, Maternal and Child Sciences , University of Verona , Verona , Italy
| | - Riccardo Rosati
- Department of Surgery , San Raffaele Hospital, San Raffaele Vita-salute University , Milan , Italy
| | | | - Daniele Marrelli
- Department of Surgery , Siena University Hospital, University of Siena , Siena , Italy
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum Surgery , Fondazione Policlinico Universitario A. Gemelli, IRCCS , Rome , Italy
| | - Antonio Macrì
- Peritoneal Surface Malignancy and Soft Tissue Sarcoma Program , University of Messina , Messina , Italy
| | - Annibale Donini
- General and Emergency Surgery , Santa Maria Della Misericordia Hospital, University of Perugia , Perugia , Italy
| | - Lorena Torroni
- Department of Diagnostic and Public Health, Unit of Epidemiology and Medical Statistics , University of Verona , Verona , Italy
| | | | - Giovanni De Manzoni
- Department and of Surgical, General and Upper GI Surgery Division, Odontostomatologic, Maternal and Child Sciences , University of Verona , Verona , Italy
| |
Collapse
|
46
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022; 28:1189-1198. [PMID: 35534566 DOI: 10.1038/s41591-022-01800-8%' and 2*3*8=6*8 and 'ika1'!='ika1%] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 01/29/2024]
Abstract
Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.
Collapse
Affiliation(s)
- Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Huiping Gao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zhen Liu
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Huamao Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jun Xiao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Hong Ma
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Wei Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zonghai Li
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
47
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022; 28:1189-1198. [PMID: 35534566 DOI: 10.1038/s41591-022-01800-8" and 2*3*8=6*8 and "sudy"="sudy] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 01/29/2024]
Abstract
Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.
Collapse
Affiliation(s)
- Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Huiping Gao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zhen Liu
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Huamao Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jun Xiao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Hong Ma
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Wei Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zonghai Li
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
48
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022; 28:1189-1198. [PMID: 35534566 DOI: 10.1038/s41591-022-01800-8����%2527%2522\'\"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 01/29/2024]
Abstract
Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.
Collapse
Affiliation(s)
- Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Huiping Gao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zhen Liu
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Huamao Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jun Xiao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Hong Ma
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Wei Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zonghai Li
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
49
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022; 28:1189-1198. [PMID: 35534566 DOI: 10.1038/s41591-022-01800-8' and 2*3*8=6*8 and 'qlv1'='qlv1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 01/29/2024]
Abstract
Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.
Collapse
Affiliation(s)
- Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Huiping Gao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zhen Liu
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Huamao Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jun Xiao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Hong Ma
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Wei Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zonghai Li
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
50
|
Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, Zhang M, Peng Z, Zhou J, Cao Y, Zhang X, Lu Z, Lu M, Yuan J, Wang Z, Wang Y, Peng X, Gao H, Liu Z, Wang H, Yuan D, Xiao J, Ma H, Wang W, Li Z, Shen L. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med 2022; 28:1189-1198. [PMID: 35534566 DOI: 10.1038/s41591-022-01800-8gje4tfn3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/25/2022] [Indexed: 01/29/2024]
Abstract
Despite success in hematologic malignancies, the treatment landscape of chimeric antigen receptor (CAR) T cell therapy for solid tumors remains limited. Claudin18.2 (CLDN18.2)-redirected CAR T cells showed promising efficacy against gastric cancer (GC) in a preclinical study. Here we report the interim analysis results of an ongoing, open-label, single-arm, phase 1 clinical trial of CLDN18.2-targeted CAR T cells (CT041) in patients with previously treated, CLDN18.2-positive digestive system cancers ( NCT03874897 ). The primary objective was safety after CT041 infusion; secondary objectives included CT041 efficacy, pharmacokinetics and immunogenicity. We treated 37 patients with one of three CT041 doses: 2.5 × 108, 3.75 × 108 or 5.0 × 108 cells. All patients experienced a grade 3 or higher hematologic toxicity. Grade 1 or 2 cytokine release syndrome (CRS) occurred in 94.6% of patients. No grade 3 or higher CRS or neurotoxicities, treatment-related deaths or dose-limiting toxicities were reported. The overall response rate (ORR) and disease control rate (DCR) reached 48.6% and 73.0%, respectively. The 6-month duration of response rate was 44.8%. In patients with GC, the ORR and DCR reached 57.1% and 75.0%, respectively, and the 6-month overall survival rate was 81.2%. These initial results suggest that CT041 has promising efficacy with an acceptable safety profile in patients with heavily pretreated, CLDN18.2-positive digestive system cancers, particularly in those with GC.
Collapse
Affiliation(s)
- Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai Ge
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhihao Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yakun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Huiping Gao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zhen Liu
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Huamao Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | | | - Jun Xiao
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Hong Ma
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Wei Wang
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Zonghai Li
- CARsgen Therapeutics Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|