1
|
Qian J, Zhang Q, Cao Y, Chu X, Gao Y, Xu H, Cai H, Wu J. Perfusion drugs for non‑muscle invasive bladder cancer (Review). Oncol Lett 2024; 27:267. [PMID: 38659423 PMCID: PMC11040539 DOI: 10.3892/ol.2024.14400] [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: 02/16/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
The high recurrence rate and poor prognosis of non-muscle invasive bladder cancer (BC) are challenges that need to be urgently addressed. Transurethral cystectomy for bladder tumors is often combined with bladder perfusion therapy, which can effectively reduce the recurrence and progression rates of BC. The present review integrated and analyzed currently available bladder perfusion drugs, mainly including chemotherapeutic agents, immunotherapeutic agents and other adjuvant perfusion drugs. Bacillus Calmette-Guerin (BCG) perfusion was the pioneering immunotherapy for early BC and still ranks high in the selection of perfusion drugs. However, BCG infusion has a high toxicity profile and has been shown to be ineffective in some patients. Due to the limitations of BCG, new bladder perfusion drugs are constantly being developed. Immunotherapeutic agents have opened a whole new chapter in the selection of therapeutic agents for bladder perfusion. The present review explored the mechanism of action, clinical dosage and adverse effects of a variety of bladder perfusion drugs currently in common use, described combined perfusion and compared the effects of certain drugs on BC.
Collapse
Affiliation(s)
- Jingyuan Qian
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Qiuchen Zhang
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Yang Cao
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Xi Chu
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Yiyang Gao
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Haifei Xu
- Department of Urology, Nantong Tumor Hospital, Nantong, Jiangsu 226006, P.R. China
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| | - Jiajia Wu
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
2
|
Zhang H, Wang J, Wu R, Zheng B, Sang Y, Wang B, Song L, Hu Y, Ma X. Self-Supplied Reactive Oxygen Species-Responsive Mitoxantrone Polyprodrug for Chemosensitization-Enhanced Chemotherapy under Moderate Hyperthermia. Adv Healthc Mater 2024; 13:e2303631. [PMID: 38278138 DOI: 10.1002/adhm.202303631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/11/2023] [Indexed: 01/28/2024]
Abstract
Currently, the secondary development and modification of clinical drugs has become one of the research priorities. Researchers have developed a variety of TME-responsive nanomedicine carriers to solve certain clinical problems. Unfortunately, endogenous stimuli such as reactive oxygen species (ROS), as an important prerequisite for effective therapeutic efficacy, are not enough to achieve the expected drug release process, therefore, it is difficult to achieve a continuous and efficient treatment process. Herein, a self-supply ROS-responsive cascade polyprodrug (PMTO) is designed. The encapsulation of the chemotherapy drug mitoxantrone (MTO) in a polymer backbone could effectively reduce systemic toxicity when transported in vivo. After PMTO is degraded by endogenous ROS of the TME, another part of the polyprodrug backbone becomes cinnamaldehyde (CA), which can further enhance intracellular ROS, thereby achieving a sustained drug release process. Meanwhile, due to the disruption of the intracellular redox environment, the efficacy of chemotherapy drugs is enhanced. Finally, the anticancer treatment efficacy is further enhanced due to the mild hyperthermia effect of PMTO. In conclusion, the designed PMTO demonstrates remarkable antitumor efficacy, effectively addressing the limitations associated with MTO.
Collapse
Affiliation(s)
- Hongjie Zhang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Jing Wang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Ruiying Wu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Benyan Zheng
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yanxiang Sang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Bibo Wang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Lei Song
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yuan Hu
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Xiaopeng Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| |
Collapse
|
3
|
Vachez E, Kefleyesus A, Bakrin N, Ranchon F, Rioufol C, Vassal O, Al-Hadeedi O, Kepenekian V, Glehen O. Eliminating the need for preoperative intravenous hyperhydration: Sodium thiosulfate as nephrotoxicity prevention in HIPEC-treated patients - A retrospective analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:107955. [PMID: 38219699 DOI: 10.1016/j.ejso.2024.107955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC) is an effective treatment for peritoneal metastases. However, HIPEC with cisplatin is associated with renal toxicity. Sodium thiosulfate (ST) has been shown to prevent cisplatin-induced toxicity. METHODS A retrospective, single-center analysis of patients treated curatively for peritoneal surface malignancy, who underwent cytoreductive surgery with cisplatin-based HIPEC between 2015 and 2020. Patients were categorized into three groups based on the management of cisplatin-induced renal toxicity: preoperative hyperhydration alone (PHH), preoperative hyperhydration with ST (PHH + ST), and ST alone. Renal function and complications, in terms of Acute (AKI) and chronic kidney injury (CKI), were monitored and analyzed during 3 postoperative months. RESULTS This study included 220 consecutive patients. Mean serum creatinine levels were 95, 57 and 61 mmol/L, for PHH, PHH + ST and ST groups, respectively (p < 0.001). Glomerular Filtration Rate (GFR) were 96, 94 and 78 ml/min/1.73 m2, respectively (p < 0.001). AKI and CKI are respectively for PHH, PHH + ST and ST groups were 21 % (n = 46), 1 % (n = 2) and 0 % vs 19 % (n = 42), 0 % and 0 % (p < 0.001), for pairwise analysis did not show any difference between PHH + ST and ST alone combination, regarding nephrological outcomes. All patients were followed 3 months postoperatively. CONCLUSION There is no need for preoperative hyperhydration when sodium-thiosulfate is used to prevent cisplatin-induced nephrotoxicity in patients undergoing cytoreductive surgery with HIPEC. These findings have implications for improving and simplifying the management of patients with peritoneal metastases undergoing HIPEC with cisplatin.
Collapse
Affiliation(s)
- Elea Vachez
- Department of Gynecology, Lyon-Sud University Hospital, Lyon, France
| | - Amaniel Kefleyesus
- Department of Surgical Oncology, Lyon-Sud University Hospital, Lyon, France; Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, Switzerland.
| | - Naoual Bakrin
- Department of Surgical Oncology, Lyon-Sud University Hospital, Lyon, France; CICLY Research Team, University Lyon 1, Lyon, France
| | - Florence Ranchon
- Unit of Oncologic Clinical Pharmacy Unit, Lyon-Sud University Hospital, Lyon, France
| | - Catherine Rioufol
- CICLY Research Team, University Lyon 1, Lyon, France; Unit of Oncologic Clinical Pharmacy Unit, Lyon-Sud University Hospital, Lyon, France
| | - Olivia Vassal
- Department of Intensive Care, Lyon-Sud University Hospital, Lyon, France
| | - Omar Al-Hadeedi
- Department of Surgical Oncology, Lyon-Sud University Hospital, Lyon, France
| | - Vahan Kepenekian
- Department of Surgical Oncology, Lyon-Sud University Hospital, Lyon, France; CICLY Research Team, University Lyon 1, Lyon, France
| | - Olivier Glehen
- Department of Surgical Oncology, Lyon-Sud University Hospital, Lyon, France; CICLY Research Team, University Lyon 1, Lyon, France
| |
Collapse
|
4
|
Bhatt A, Glehen O, Zivanovic O, Brennan D, Nadeau C, Van Driel W, Bakrin N. The 2022 PSOGI International Consensus on HIPEC Regimens for Peritoneal Malignancies: Epithelial Ovarian Cancer. Ann Surg Oncol 2023; 30:8115-8137. [PMID: 37561343 DOI: 10.1245/s10434-023-13932-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/27/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND AND AIM We report the results of an international consensus on hyperthermic intraperitoneal chemotherapy (HIPEC) regimens for epithelial ovarian cancer (EOC) performed with the following goals: To define the indications for HIPEC To identify the most suitable HIPEC regimens for each indication in EOC To identify areas of future research on HIPEC To provide recommendations for some aspects of perioperative care for HIPEC METHODS: The Delphi technique was used with two rounds of voting. There were three categories of questions: evidence-based recommendations [using the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) system with the patient, intervention, comparator, and outcome (PICO) method], an opinion survey, and research recommendations. RESULTS Seventy-three (67.5%) of 108 invited experts responded in round I, and 68 (62.9%) in round II. Consensus was achieved for 34/38 (94.7%) questions. However, a strong positive consensus that would lead to inclusion in routine care was reached for only 6/38 (15.7%) questions. HIPEC in addition to interval cytoreductive surgery (CRS) received a strong positive recommendation that merits inclusion in routine care. Single-agent cisplatin was the only drug recommended for routine care, and OVHIPEC-1 was the most preferred regimen. The panel recommended performing HIPEC for a minimum of 60 min with a recommended minimum intraabdominal temperature of 41°C. Nephroprotection with sodium thiosulfate should be used for cisplatin HIPEC. CONCLUSIONS The results of this consensus should guide clinical decisions on indications of HIPEC and the choice and various parameters of HIPEC regimens and could fill current knowledge gaps. These outcomes should be the basis for designing future clinical trials on HIPEC in EOC.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, KD Hospital, Ahmedabad, India.
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier, Lyon-sud, Lyon, France
| | - Oliver Zivanovic
- Department of Gynecological Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donal Brennan
- UCD Gynaecological Oncology Group, UCD School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Cedric Nadeau
- Department of Gynecological Oncology, CHU de Poitiers, Poitiers, Cedex, France
| | - Willemien Van Driel
- Department of Gynecological Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Naoual Bakrin
- Department of Surgical Oncology, Centre Hospitalier, Lyon-sud, Lyon, France
| |
Collapse
|
5
|
Hashemi M, abbasiazam A, Oraee-Yazdani S, Lenzer J. Response of human glioblastoma cells to hyperthermia: Cellular apoptosis and molecular events. Tissue Cell 2022; 75:101751. [DOI: 10.1016/j.tice.2022.101751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
|
6
|
Photo-induced processes of iron oxide nanoparticles to enhance laser therapy. BIOMEDICAL PHOTONICS 2022. [DOI: 10.24931/2413-9432-2021-10-4-44-58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nanoparticles are used as drug carriers to increase the selectivity and effectiveness of therapy, as well as for combined therapy that utilizes different effects. Iron oxide nanoparticles are promising in this aspect. Due to magnetic properties, they can be used as a contrast agent for magnetic resonance imaging. Also, iron oxide nanoparticles could be coated with a photosensitizer for photodynamic therapy and their laser or magnetic heating can be used for phototherapy. Local enhancement of the electromagnetic field near iron oxide nanoparticles can increase the fluorescence intensity of photosensitizers and the efficiency of singlet oxygen generation. This paper presents the results of a study of iron oxide nanoparticles focused on the photophysical aspects of the formation of “hot spots” under laser irradiation. The photoinduced effects of iron oxide nanoparticles observed in in vitro experiments lead to the rupture of lysosomes. Theoretical modeling showed that the heating of iron oxide nanoparticles with a radius of 35 nm under the action of laser radiation is about 89°C and 19°C for wavelengths of 458 and 561 nm, respectively. Local field enhancement occurs in pairs of nanoparticles of various sizes and strongly depends on the distance between them. The maximum gain is achieved at small distances between nanoparticles. For a dimer of nanoparticles with radii of 10 and 35 nm at a distance of 1 nm, an enhancement factor of two orders of magnitude was obtained. The investigated phenomenon of «hot spots» is in demand for precision therapy, because the photo-induced processes occur at small distances between nanoparticles, in areas of their high accumulation.
Collapse
|
7
|
Fung X, Li I, Chandrakumaran K, Cecil T, Dayal S, Tzivanakis A, Moran B, Mohamed F. Early postoperative intraperitoneal chemotherapy (EPIC) following cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) in 632 patients with pseudomyxoma peritonei of appendiceal origin: A single institution experience. Eur J Surg Oncol 2022; 48:1614-1618. [DOI: 10.1016/j.ejso.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/14/2022] [Accepted: 02/01/2022] [Indexed: 11/28/2022] Open
|
8
|
Lee WT, Yoon J, Kim SS, Kim H, Nguyen NT, Le XT, Lee ES, Oh KT, Choi HG, Youn YS. Combined Antitumor Therapy Using In Situ Injectable Hydrogels Formulated with Albumin Nanoparticles Containing Indocyanine Green, Chlorin e6, and Perfluorocarbon in Hypoxic Tumors. Pharmaceutics 2022; 14:pharmaceutics14010148. [PMID: 35057044 PMCID: PMC8781012 DOI: 10.3390/pharmaceutics14010148] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/28/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Combined therapy using photothermal and photodynamic treatments together with chemotherapeutic agents is considered one of the most synergistic treatment protocols to ablate hypoxic tumors. Herein, we sought to fabricate an in situ-injectable PEG hydrogel system having such multifunctional effects. This PEG hydrogel was prepared with (i) nabTM-technique-based paclitaxel (PTX)-bound albumin nanoparticles with chlorin-e6 (Ce6)-conjugated bovine serum albumin (BSA-Ce6) and indocyanine green (ICG), named ICG/PTX/BSA-Ce6-NPs (~175 nm), and (ii) an albumin-stabilized perfluorocarbon (PFC) nano-emulsion (BSA-PFC-NEs; ~320 nm). This multifunctional PEG hydrogel induced moderate and severe hyperthermia (41−42 °C and >48 °C, respectively) at the target site under two different 808 nm laser irradiation protocols, and also induced efficient singlet oxygen (1O2) generation under 660 nm laser irradiation supplemented by oxygen produced by ultrasound-triggered PFC. Due to such multifunctionality, our PEG hydrogel formula displayed significantly enhanced killing of three-dimensional 4T1 cell spheroids and also suppressed the growth of xenografted 4T1 cell tumors in mice (tumor volume: 47.7 ± 11.6 and 63.4 ± 13.0 mm3 for photothermal and photodynamic treatment, respectively, vs. PBS group (805.9 ± 138.5 mm3), presumably based on sufficient generation of moderate heat as well as 1O2/O2 even under hypoxic conditions. Our PEG hydrogel formula also showed excellent hyperthermal efficacy (>50 °C), ablating the 4T1 tumors when the irradiation duration was extended and output intensity was increased. We expect that our multifunctional PEG hydrogel formula will become a prototype for ablation of otherwise poorly responsive hypoxic tumors.
Collapse
Affiliation(s)
- Woo Tak Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Johyun Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Sung Soo Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Hanju Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Nguyen Thi Nguyen
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Xuan Thien Le
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
| | - Eun Seong Lee
- Department of Biotechnology and Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si 14662, Gyeonggi-do, Korea;
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Gyeonggi-do, Korea;
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (W.T.L.); (J.Y.); (S.S.K.); (H.K.); (N.T.N.); (X.T.L.)
- Correspondence: ; Tel.: +82-31-290-7785
| |
Collapse
|
9
|
Zheng N, Xu A, Lin X, Mo Z, Xie X, Huang Z, Liang Y, Cai Z, Tan J, Shao X. Whole-body hyperthermia combined with chemotherapy and intensity-modulated radiotherapy for treatment of advanced nasopharyngeal carcinoma: a retrospective study with propensity score matching. Int J Hyperthermia 2021; 38:1304-1312. [PMID: 34468276 DOI: 10.1080/02656736.2021.1971778] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Several studies have reported the combination of intracavity or cervical lymph node hyperthermia with chemoradiotherapy (CRT) to improve clinical outcomes in nasopharyngeal carcinoma (NPC), but the combination with whole-body hyperthermia (WBH) for treating NPC is unexplored. We aimed to assess the efficacy of the combination of radiotherapy, chemotherapy and WBH in patients with locoregionally advanced NPC. METHODS Between July 2008 and November 2012, 239 newly diagnosed NPC patients were enrolled in a pre-propensity score-matched cohort, including 193 patients who received CRT (CRT group) and 46 who underwent CRT with WBH (HCRT group). The feasibility and clinical outcomes of both groups were evaluated and toxicities assessed. Survival rates were assessed using the Kaplan-Meier method, log-rank test and Cox regression. RESULTS Following propensity score matching, 46 patients from each group were included. The 5-year overall survival (OS) rates were 65.2% in the CRT group and 80.3% in the HCRT group (p=.027). In contrast, the other survival outcomes at 5 years were similar between the groups: locoregional recurrence-free survival (LRRFS), 74.7% vs. 87.6% (p=.152); distant metastasis-free survival (DMFS), 67.4% vs. 77.9% (p=.125); and progression-free survival (PFS), 53.1% vs. 69.2% (p=.115). In the multivariate analyses, the only two independent predictors of OS were clinical stage and HCRT. CONCLUSIONS These results suggest that WBH, when combined with CRT, can improve the OS of patients with advanced NPC.
Collapse
Affiliation(s)
- Naiying Zheng
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Anan Xu
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Xiantao Lin
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, PR China
| | - Zhiwen Mo
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Xiaoxue Xie
- Department of Radiotherapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, PR China
| | - Zhong Huang
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Ying Liang
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Zhihua Cai
- Department of Chemotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Jianming Tan
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| | - Xunfan Shao
- Department of Radiotherapy, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
10
|
Hannon G, Tansi FL, Hilger I, Prina‐Mello A. The Effects of Localized Heat on the Hallmarks of Cancer. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gary Hannon
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
| | - Felista L. Tansi
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology Jena University Hospital—Friedrich Schiller University Jena Am Klinikum 1 07740 Jena Germany
| | - Adriele Prina‐Mello
- Nanomedicine and Molecular Imaging Group Trinity Translational Medicine Institute Dublin 8 Ireland
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute Trinity College Dublin Dublin 8 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute Trinity College Dublin Dublin 2 Ireland
| |
Collapse
|
11
|
Bhatt A, de Hingh I, Van Der Speeten K, Hubner M, Deraco M, Bakrin N, Villeneuve L, Kusamura S, Glehen O. HIPEC Methodology and Regimens: The Need for an Expert Consensus. Ann Surg Oncol 2021; 28:9098-9113. [PMID: 34142293 DOI: 10.1245/s10434-021-10193-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hyperthermic intraperitoneal chemotherapy (HIPEC) is performed with a wide variation in methodology, drugs, and other elements vital to the procedure. Adoption of a limited number of regimens could increase the collective experience of peritoneal oncologists, make comparison between studies more meaningful, and lead to a greater acceptance of results from randomized trials. This study aimed to determine the possibility of standardizing HIPEC methodology and regimens and to identify the best method of performing such a standardization. METHODS A critical review of preclinical and clinical studies evaluating the pharmacokinetic aspects of different HIPEC drugs and drug regimens, the impact of hyperthermia, and the efficacy of various HIPEC regimens as well as studies comparing different regimens was performed. RESULTS The preclinical and clinical data were limited, and studies comparing different regimens were scarce. Many of the regimens were neither supported by preclinical rationale or data nor validated by a dose-escalating formal phase 1 trial. All the regimens were based on pharmacokinetic data and did not take chemosensitivity of peritoneal metastases into account. Personalized medicine approaches such as patient-derived tumor organoids could offer a solution to this problem, although clinical validation is likely to be challenging. CONCLUSIONS Apart from randomized trials, more translational research and phases 1 and 2 studies are needed. While waiting for better preclinical and clinical evidence, the best way to minimize heterogeneity is by an expert consensus that aims to identify and define a limited number of regimens for each indication and primary site. The choice of regimen then can be tailored to the patient profile and its expected toxicity and the methodology according regional factors.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, Zydus Hospital, Ahmedabad, India
| | - Ignace de Hingh
- Department of Surgical Oncology, Catharina Hospital, Eidhoven, The Netherlands
| | | | - Martin Hubner
- Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Marcello Deraco
- Department of Surgical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Naoual Bakrin
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| | - Laurent Villeneuve
- Department of Clinical Research, Hospices Civils de Lyon, Centre Hospitalier Lyon-sud, Lyon, France
| | - Shigeki Kusamura
- Department of Surgical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France.
| |
Collapse
|
12
|
Reutovich MY, Krasko OV, Sukonko OG. Hyperthermic intraperitoneal chemotherapy in prevention of gastric cancer metachronous peritoneal metastases: a systematic review. J Gastrointest Oncol 2021; 12:S5-S17. [PMID: 33968422 DOI: 10.21037/jgo-20-129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer progression resulting in metachronous peritoneal metastasizing is almost always associated with an adverse prognosis. This review discusses various options of preventing metachronous peritoneal metastases in radically operated gastric cancer patients. Also examined are different hyperthermic intraperitoneal chemotherapy (HIPEC) regimens employed in gastric cancer treatment, postoperative morbidity and mortality rates and long-term treatment outcomes. The authors also review their own experience of using HIPEC based on the combination of cisplatin and doxorubicin in doses of 50 mg/m2 at 42 °C for 1 h to prevent gastric cancer peritoneal dissemination. As a result, progression-free survival rose from 19.6%±5.6% to 47.1%±6.3% (Plog-rank <0.001) and dissemination-free survival-from 22.7%±6.0% to 51.9%±6.3% (Plog-rank <0.001). It is noted that the combination of the described HIPEC regimen with systemic chemotherapy helped raise metastases-free 3-year survival rate to up to 91.0%±9.0% (Plog-rank =0.025) compared with 48.6%±6.4% for patients who underwent only a combined surgery/HIPEC treatment. HIPEC is a promising combined treatment strategy for radically operated gastric cancer patients that can improve patient survival and decrease peritoneal dissemination rate. However, the number of randomized studies on adjuvant HIPEC are still insufficient for a subgroup assessment of efficacy of the given chemotherapy regimens and generation of evidence-based recommendations on the individual use of chemotherapy agents and their combinations, and HIPEC procedural techniques. Further prospective randomized studies are needed to assess the practicability of complementing HIPEC with adjuvant systemic chemotherapies.
Collapse
Affiliation(s)
- Mikhail Yu Reutovich
- Gastroesophageal Pathology Department, N.N. Alexandrov National Cancer Center, Minsk, Belarus
| | - Olga V Krasko
- United Institute of Informatics Problems, National Academy of Sciences, Minsk, Belarus
| | - Oleg G Sukonko
- Gastroesophageal Pathology Department, N.N. Alexandrov National Cancer Center, Minsk, Belarus
| |
Collapse
|
13
|
Fan B, Bu Z, Zhang J, Zong X, Ji X, Fu T, Jia Z, Zhang Y, Wu X. Phase II trial of prophylactic hyperthermic intraperitoneal chemotherapy in patients with locally advanced gastric cancer after curative surgery. BMC Cancer 2021; 21:216. [PMID: 33653317 PMCID: PMC7923597 DOI: 10.1186/s12885-021-07925-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background HIPEC is an emerging procedure to treat peritoneal metastasis of gastric cancer. Data about HIPEC in locally advanced gastric cancer is scarce. The purpose of this trial is to evaluate the safety and toxicity of prophylactic HIPEC with cisplatin for patients with locally advanced gastric cancer. Methods From March 2015 to November 2016, a prospective, randomized phase II trial was conducted. After radical gastrectomy, patients in the experimental group underwent HIPEC with cisplatin followed by adjuvant chemotherapy with SOX regime. Patients in the other group were treated with SOX regime alone. Postoperative complications and patient survival were compared. Results In total, 50 patients were eligible for analyses. No significant difference was found in the incidence of postoperative complications including anastomotic/intestinal leakage, liver dysfunction, bone marrow suppression, wound infection and ileus (P > 0.05). Mean duration of hospitalization after radical gastrectomy was 11.7 days. 12.2 days in experimental group and 10.8 days in control group respectively (P = 0.255). The percentage of patients with elevated tumor markers was 12.1% in experimental group, which was significantly lower than 41.2% in control group (P = 0.02). 3-year RFS of patients who treated with or without prophylactic HIPEC were 84.8 and 88.2% respectively (P = 0.986). In the multivariate analysis, pathological T stage was the only independent risk factor for the RFS of patients (P = 0.012, HR =15.071). Conclusion Additional intraoperative HIPEC with cisplatin did not increase postoperative complications for locally advanced gastric cancer after curative surgery. Prophylactic HIPEC with cisplatin was safe and tolerable, while it did not reduce the risk of peritoneal recurrence in this trial, supporting further studies to validate the efficacy of it. Trial registration Chinese Clinical Trial Registry, ChiCTR2000038331. Registered 18 September 2020 - Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=59692.
Collapse
Affiliation(s)
- Biao Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Zhaode Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ji Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xianglong Zong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xin Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Tao Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Ziyu Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Yinan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China
| | - Xiaojiang Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer center, Peking University Cancer Hospital and Institute, Fu-Cheng Road 52#, Hai-Dian District, Beijing, 100142, China.
| |
Collapse
|
14
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
15
|
Wen H, Tamarov K, Happonen E, Lehto V, Xu W. Inorganic Nanomaterials for Photothermal‐Based Cancer Theranostics. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000207] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Huang Wen
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Konstantin Tamarov
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Emilia Happonen
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Vesa‐Pekka Lehto
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Wujun Xu
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| |
Collapse
|
16
|
Comparison of Iron Oxide Nanoparticles in Photothermia and Magnetic Hyperthermia: Effects of Clustering and Silica Encapsulation on Nanoparticles’ Heating Yield. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10207322] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photothermal therapy is gathering momentum. In order to assess the effects of the encapsulation of individual or clustered superparamagnetic iron oxide nanoparticles (SPIONs) on nanoparticle light-to-heat conversion, we designed and tested individual and clustered SPIONs encapsulated within a silica shell. Our study compared both photothermia and magnetic hyperthermia, and it involved individual SPIONs as well as silica-encapsulated individual and clustered SPIONs. While, as expected, SPION clustering reduced heat generation in magnetic hyperthermia, the silica shell improved SPION heating in photothermia.
Collapse
|
17
|
Sugarbaker PH, Stuart OA. Perioperative FOLFOX in management of peritoneal metastases of colorectal cancer. Case report of 2 patients. Int J Surg Case Rep 2020; 75:279-285. [PMID: 32979826 PMCID: PMC7519275 DOI: 10.1016/j.ijscr.2020.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 01/13/2023] Open
Abstract
Peritoneal metastases from colorectal cancer are lethal. Cytoreductive surgery plus perioperative chemotherapy is a treatment option. A perioperative FOLFOX regimen with intraperitoneal oxaliplatin was described. The pharmacokinetic studies suggested efficacy.
Introduction Colorectal cancer can disseminate malignant cells to the peritoneal surfaces which over time progress to peritoneal metastases. Management of this type of metastatic disease has been approached using a combined treatment that consists of cytoreductive surgery and perioperative chemotherapy. To optimize these treatments a more effective chemotherapy that is used as planned part of the surgical procedure is required. Presentation of cases Pharmacologic studies to initiate a new perioperative chemotherapy treatment were modeled after the successful systemic treatments of metastatic colorectal cancer referred to as FOLFOX. A management plan that included all of the essential features of successful systemic chemotherapy was formulated. Pharmacokinetic studies of 5-fluorouracil given both intravenously and intraperitoneally and oxaliplatin given intraperitoneally were investigated. Discussion The compatibility of 5-fluorouracil and oxaliplatin was documented showing no degradation of either drug when they were mixed in-vitro over 24 h. The pharmacokinetic analysis of hyperthermic intraperitoneal chemotherapy (HIPEC) with oxaliplatin showed cytotoxic concentrations for 120 min. In order to maximize oxaliplatin’s effect, both intravenous bolus and continuous infusion 5-fluorouracil was combined with early postoperative intraperitoneal chemotherapy (EPIC). In total, 200 mg/m2 of oxaliplatin was combined with a minimum of 1600 mg of 5-fluorouracil over the 24 -h treatment plan. Conclusion This perioperative FOLFOX treatment was completed in 2 patients and the clinical effectiveness as a result of this in-depth case reports was presented. Formal phase II studies, as a result of these pharmacokinetic and clinical investigations, have been initiated.
Collapse
Affiliation(s)
- Paul H Sugarbaker
- Program in Peritoneal Surface Malignancies, MedStar Washington Hospital Center, Washington, DC, USA.
| | - O Anthony Stuart
- Program in Peritoneal Surface Malignancies, MedStar Washington Hospital Center, Washington, DC, USA
| |
Collapse
|
18
|
Yonemura Y, Iahibashi H, Sako S, Mizumoto A, Takao N, Ichinose M, Motoi S, Liu Y, Wakama S, Kamada Y, Nishihara K. Advances with pharmacotherapy for peritoneal metastasis. Expert Opin Pharmacother 2020; 21:2057-2066. [PMID: 32783786 DOI: 10.1080/14656566.2020.1793957] [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: 10/23/2022]
Abstract
INTRODUCTION A new treatment strategy involving cytoreductive surgery (CRS) combined with perioperative intraperitoneal (IP) chemotherapy was proposed in 1999 by the Peritoneal Surface Oncology Group International, and the strategy is now justified as a state-of-the-art treatment to improve the long-term survival of patients with peritoneal metastasis (PM). To achieve cure in the patients with PM, complete removal of macroscopic tumors and eradication of micrometastasis on the peritoneum, left after CRS are essential. Systemic chemotherapy is not indicated for the treatment of PM. In contrast, intraperitoneal (IP) chemotherapy brings about significantly higher locoregional dose intensity in the peritoneal cavity and subperitoneal tissues. In combination with anticancer drugs, hyperthermia enhances cytotoxicity against cancer cells. AREA COVERED This article provides a systematic overview of PM from various cancers including gastric, colorectal, small bowel, appendiceal cancer, and mesothelioma. It also includes all the essential aspects of therapy. EXPERT OPINION CRS plus perioperative intraperitoneal chemotherapy is safe with acceptable morbidity and mortality. It is justified as a standard treatment to improve the long-term survival of patients with PM and is now performed with curative intent for PM from various malignancies.
Collapse
Affiliation(s)
- Yutaka Yonemura
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan.,Department of Peritoneal Surface Malignancy Treatment, Kusatsu General Hospital , Kusatsu City, Shiga, Japan
| | - Haruaki Iahibashi
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| | - Shouzou Sako
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| | - Akiyoshi Mizumoto
- Department of Peritoneal Surface Malignancy Treatment, Kusatsu General Hospital , Kusatsu City, Shiga, Japan
| | - Nobuyuki Takao
- Department of Peritoneal Surface Malignancy Treatment, Kusatsu General Hospital , Kusatsu City, Shiga, Japan
| | - Masumi Ichinose
- Department of Peritoneal Surface Malignancy Treatment, Kusatsu General Hospital , Kusatsu City, Shiga, Japan
| | - Shunsuke Motoi
- Department of Peritoneal Surface Malignancy Treatment, Kusatsu General Hospital , Kusatsu City, Shiga, Japan
| | - Yang Liu
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| | - Satoshi Wakama
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| | - Yasuyuki Kamada
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| | - Kazurou Nishihara
- Department of Regional Cancer Therapies, Peritoneal Surface Malignancy Treatment Center, Kishiwada Tokushukai Hospital , Kishiwada City, Oosaka-Fu, Japan
| |
Collapse
|
19
|
Fabijańska M, Orzechowska M, Rybarczyk-Pirek AJ, Dominikowska J, Bieńkowska A, Małecki M, Ochocki J. Simple Trans-Platinum Complex Bearing 3-Aminoflavone Ligand Could Be a Useful Drug: Structure-Activity Relationship of Platinum Complex in Comparison with Cisplatin. Int J Mol Sci 2020; 21:ijms21062116. [PMID: 32204470 PMCID: PMC7139614 DOI: 10.3390/ijms21062116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 01/05/2023] Open
Abstract
Following previous studies devoted to trans–Pt(3-af)2Cl2, in this paper, the molecular structure and intermolecular interactions of the title complex are compared with other cisplatin analogues of which the crystal structures are presented in the Cambridge Structural Database (CSD). Molecular Hirshfeld surface analysis and computational methods were used to examine a possible relationship between the structure and anticancer activity of trans–Pt(3-af)2Cl2. The purpose of the article was also to investigate the effect of hyperthermia on the anticancer activity of cisplatin, cytostatics used in the treatment of patients with ovarian cancer and a new analogue of cisplatin-trans–Pt(3-af)2Cl2. The study was conducted on two cell lines of ovarian cancer sensitive to Caov-3 cytostatics and the OVCAR-3 resistant cisplatin line. The study used the MTT (3-(4,5-dimethylthiazol-2,5-diphenyltetrazolium bromide) cell viability assay, LDH (lactate dehydrogenase), and the quantitative evaluation method for measuring gene expression, i.e., qPCR with TagMan probes. Reduced survivability of OVCAR-3 and Caov-3 cells exposed to cytostatics at elevated temperatures (37 °C, 40 °C, 43 °C) was observed. Hyperthermia may increase the sensitivity of cells to platinum-based antineoplastic drugs and paclitaxel, which may be associated with the reduction of gene expression related to apoptotic processes.
Collapse
Affiliation(s)
- Małgorzata Fabijańska
- Department of Bioinorganic Chemistry, Medical University of Lodz, 1 Muszynskiego St., 90-151 Łódź, Poland
- Correspondence: (M.F.); (J.O.); Tel.: +48-(42)-6779220 (J.O.)
| | - Magdalena Orzechowska
- Department of Applied Pharmacy, Medical University of Warsaw, 1 Banacha St., 02–097 Warsaw, Poland; (M.O.); (A.B.); (M.M.)
| | - Agnieszka J. Rybarczyk-Pirek
- Theoretical and Structural Chemistry Group, Department of Physical Chemistry, Faculty of Chemistry, University of Łódź, Pomorska 163/165, 90-236 Łódź, Poland; (A.J.R.-P.); (J.D.)
| | - Justyna Dominikowska
- Theoretical and Structural Chemistry Group, Department of Physical Chemistry, Faculty of Chemistry, University of Łódź, Pomorska 163/165, 90-236 Łódź, Poland; (A.J.R.-P.); (J.D.)
| | - Alicja Bieńkowska
- Department of Applied Pharmacy, Medical University of Warsaw, 1 Banacha St., 02–097 Warsaw, Poland; (M.O.); (A.B.); (M.M.)
| | - Maciej Małecki
- Department of Applied Pharmacy, Medical University of Warsaw, 1 Banacha St., 02–097 Warsaw, Poland; (M.O.); (A.B.); (M.M.)
| | - Justyn Ochocki
- Department of Bioinorganic Chemistry, Medical University of Lodz, 1 Muszynskiego St., 90-151 Łódź, Poland
- Correspondence: (M.F.); (J.O.); Tel.: +48-(42)-6779220 (J.O.)
| |
Collapse
|
20
|
Jun HJ, Park SJ, Kang HJ, Lee GY, Lee N, Park JH, Yoo HS. The Survival Benefit of Combination Therapy With Mild Temperature Hyperthermia and an Herbal Prescription of Gun-Chil-Jung in 54 Cancer Patients Treated With Chemotherapy or Radiation Therapy: A Retrospective Study. Integr Cancer Ther 2020; 19:1534735420926583. [PMID: 32449629 PMCID: PMC7249570 DOI: 10.1177/1534735420926583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/25/2020] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
Abstract
Background: The combination of herbal medicine with conventional treatment increases the survival rate of cancer patients, but the effect is not great. Hyperthermia may have a synergistic effect with herbal medicine alongside conventional medicine. Objective: To monitor the efficacy of hyperthermia together with Gun-Chil-Jung (GCJ) capsule for event-free survival (EFS) and overall survival (OS) for the treatment of various cancers. Methods: We collected data retrospectively on 54 cancer patients of all stages. They were divided into 4 groups according to each hyperthermia or GCJ treatment period. Hyperthermia with 0.46 MHz radiofrequency wave was applied a power of 50 to 100 W for 70 minutes. GCJ capsules were administered orally 3 times a day. Results: The median follow-up was 13.4 months, and 25 (55.6%) patients showed disease-related events. Hyperthermia with GCJ treatment was administered in combination group (n = 36, 66.7%) and traditional Korean medicine-only group (n = 17, 31.5%). The median EFS was 190 days, and the median OS was 390 days. The group of hyperthermia 7 times or fewer and GCJ more than 28 days showed longer EFS and OS. The analysis of superiority between hyperthermia and GCJ showed no significant difference (EFS, P = .55; OS, P = .364). Conclusions: The combination of hyperthermia 1 to 2 times a week with GCJ treatment may improve survival of cancer patients treated or being treated with conventional cancer therapies.
Collapse
Affiliation(s)
- Hyeong Joon Jun
- Seoul Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
| | - So-Jung Park
- Dunsan Korean Medicine Hospital of
Daejeon University, Daejeon, Republic of Korea
| | - Hwi-Joong Kang
- Seoul Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
| | - Ga-Young Lee
- Cheonan Korean Medicine Hospital of
Daejeon University, Cheonan, Republic of Korea
| | - Namhun Lee
- Cheonan Korean Medicine Hospital of
Daejeon University, Cheonan, Republic of Korea
| | - Ji Hye Park
- Seoul Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
| | - Hwa-Seung Yoo
- Seoul Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Wang L, Hervault A, Southern P, Sandre O, Couillaud F, Thanh NTK. In vitro exploration of the synergistic effect of alternating magnetic field mediated thermo–chemotherapy with doxorubicin loaded dual pH- and thermo-responsive magnetic nanocomposite carriers. J Mater Chem B 2020; 8:10527-10539. [DOI: 10.1039/d0tb01983f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticle induced hyperthermia has been considered as a promising approach for cancer treatment for decades.
Collapse
Affiliation(s)
- Lilin Wang
- Biophysics Group
- Department of Physics & Astronomy
- University College London
- London
- UK
| | - Aziliz Hervault
- Biophysics Group
- Department of Physics & Astronomy
- University College London
- London
- UK
| | - Paul Southern
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories
- London
- UK
- Department of Medical Physics and Biomedical Engineering
- University College London
| | - Olivier Sandre
- Laboratoire de Chimie des Polymères Organiques (LCPO)
- Univ. Bordeaux
- CNRS
- Bordeaux INP
- UMR 5629
| | - Franck Couillaud
- Molecular Imaging and Innovative Therapies (IMOTION)
- Univ. Bordeaux
- EA7435
- Bordeaux
- France
| | - Nguyen Thi Kim Thanh
- Biophysics Group
- Department of Physics & Astronomy
- University College London
- London
- UK
| |
Collapse
|
22
|
Radiofrequency deep hyperthermia combined with chemotherapy in the treatment of advanced non-small cell lung cancer. Chin Med J (Engl) 2019; 132:922-927. [PMID: 30958433 PMCID: PMC6595762 DOI: 10.1097/cm9.0000000000000156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background In the era of precision medicine, chemotherapy is still considered the cornerstone of treatment for lung cancer patients without gene mutations. How to reduce the toxicity and increase the efficiency of chemotherapy is worth exploring. This study aimed to investigate the curative effects and safety of hyperthermia combined with chemotherapy (HCT) for advanced patients with non-small cell lung cancer (NSCLC), especially those with malignant pleural effusion. Methods We retrospectively evaluated medical records of 93 patients with advanced NSCLC (stage IIIB-IV) from March 2011 to January 2014. The patients were divided into HCT and chemotherapy (CT) groups. The HCT group was treated with gemcitabine and cisplatin (GP) regimen combined with regional radiofrequency deep hyperthermia, while the CT group was treated with GP regimen only. Those with malignant pleural effusion extra underwent thoracentesis and intrapleural injection chemotherapy combined with hyperthermic or not. Clinical treatment results and adverse reactions were compared and analyzed after treatment. SPSS 19.0 software (SPSS Inc., USA) was used for statistical data processing. P values less than 0.05 were accepted to be statistically significant. Results Among the 93 patients, HCT group included 48 patients (16 patients with malignant pleural effusion), CT group included 45 patients (10 patients with malignant pleural effusion). There was no significant difference between the two groups in patient characteristics. The overall response rate (ORR) of pleural effusions was much better in HCT group than that in CT group (81.2% vs. 40.0%, P = 0.046). The patients in HCT group had lower incidence rate of weakness (12.5% vs. 46.7%, χ2 = 13.16, P < 0.001) and gastrointestinal (25.0% vs. 77.8%, χ2 = 25.88, P < 0.001) adverse reactions than that in CT group. The objective tumor response and survival showed no significant differences. Conclusions Hyperthermia combined with chemotherapy might lead to the development of better therapeutic strategy for advanced NSCLC with malignant pleural effusion patients. Also, it could greatly reduce the chemotherapy toxic effects in the incidence of weakness and gastrointestinal adverse reactions in advanced NSCLC patients.
Collapse
|
23
|
Photocracking Silica: Tuning the Plasmonic Photothermal Degradation of Mesoporous Silica Encapsulating Gold Nanoparticles for Cargo Release. INORGANICS 2019. [DOI: 10.3390/inorganics7060072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The degradation of bionanomaterials is essential for medical applications of nanoformulations, but most inorganic-based delivery agents do not biodegrade at controllable rates. In this contribution, we describe the controllable plasmonic photocracking of gold@silica nanoparticles by tuning the power and wavelength of the laser irradiation, or by tuning the size of the encapsulated gold cores. Particles were literally broken to pieces or dissolved from the inside out upon laser excitation of the plasmonic cores. The photothermal cracking of silica, probably analogous to thermal fracturing in glass, was then harnessed to release cargo molecules from gold@silica@polycaprolactone nanovectors. This unique and controllable plasmonic photodegradation has implications for nanomedicine, photopatterning, and sensing applications.
Collapse
|
24
|
Lu CH, Chen WT, Hsieh CH, Kuo YY, Chao CY. Thermal cycling-hyperthermia in combination with polyphenols, epigallocatechin gallate and chlorogenic acid, exerts synergistic anticancer effect against human pancreatic cancer PANC-1 cells. PLoS One 2019; 14:e0217676. [PMID: 31150487 PMCID: PMC6544372 DOI: 10.1371/journal.pone.0217676] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Hyperthermia (HT) has shown feasibility and potency as an anticancer therapy. Administration of HT in the chemotherapy has previously enhanced the cytotoxicity of drugs against pancreatic cancer. However, the drugs used when conducting these studies are substantially conventional chemotherapeutic agents that may cause unwanted side effects. Additionally, the thermal dosage in the treatment of cancer cells could also probably harm the healthy cells. The purpose of this work was to investigate the potential of the two natural polyphenolic compounds, epigallocatechin gallate (EGCG) and chlorogenic acid (CGA), as heat synergizers in the thermal treatment of the PANC-1 cells. Furthermore, we have introduced a unique strategy entitled the thermal cycling-hyperthermia (TC-HT) that is capable of providing a maximum synergy and minimal side effect with the anticancer compounds. Our results demonstrate that the combination of the TC-HT and the CGA or EGCG markedly exerts the anticancer effect against the PANC-1 cells, while none of the single treatment induced such changes. The synergistic activity was attributed to the cell cycle arrest at the G2/M phase and the induction of the ROS-dependent mitochondria-mediated apoptosis. These findings not only represent the first in vitro thermal synergistic study of natural compounds in the treatment of pancreatic cancer, but also highlight the potential of the TC-HT as an alternative strategy in thermal treatment.
Collapse
Affiliation(s)
- Chueh-Hsuan Lu
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Biomedical & Molecular Imaging Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Ting Chen
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Biomedical & Molecular Imaging Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Hsiung Hsieh
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Biomedical & Molecular Imaging Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Yi Kuo
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Biomedical & Molecular Imaging Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Yu Chao
- Department of Physics, Lab for Medical Physics & Biomedical Engineering, National Taiwan University, Taipei, Taiwan
- Biomedical & Molecular Imaging Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Institute of Applied Physics, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
25
|
He M, Sun J, Zhao D, He H, Wang B, Xu L, Shang Y, Ren S, Zhang Y, Wu T. Modified-FOLFIRINOX combined with deep regional hyperthermia in pancreatic cancer: a retrospective study in Chinese patients. Int J Hyperthermia 2019; 36:394-402. [PMID: 30917701 DOI: 10.1080/02656736.2019.1579371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND FOLFIRINOX chemotherapy displays significant survival improvements in patients with pancreatic cancer. However, toxicities have hampered enthusiasm for the use of FOLFIRINOX in full dose. In order to increase the tolerability, many researchers focused on the modification of FOLFIRINOX. On the other hand, hyperthermia (HT) has been considered as an effective ancillary treatment for cancer therapy. Up to now, there is no report evaluating combining deep regional hyperthermia (DRHT) with modified-FOLFIRINOX for pancreatic cancer patients. METHODS In this study, we conducted a retrospective review of pancreatic cancer patients treated with the combination of new form modified-FOLFIRINOX and DRHT (BSD2000). Patients underwent chemotherapy that included low-dose irinotecan (70-130 mg/m2), oxaliplatin (65-70 mg/m2) on day 1 and 5-FU (2400 mg/m2 as a 46 h continuous infusion, no bolus) or capecitabine (CAP) (1000 mg/m2 twice daily on days 1-10) or tegafur, gimeracil and oteracil potassium (TS-1) (80-120 mg/d twice daily on days 1-10), 2-week schedule. Generally, DRHT treatment was performed weekly, 45 min for each time during chemotherapy. RESULTS The patients receiving mFOLFIRINOX as the first line chemotherapy combining with DRHT, obtained an improvement in OS and PFS, 17 months (95% CI 1.97-32.03 months) and 4 months (95% CI 0-8.29 months) respectively. Overall, this combination regimen was safe; 17.6% patients suffered from grade 3/4 toxicities. CONCLUSIONS In conclusion, we conducted a retrospective study combining mFOLFIRINOX and DRHT, which was well tolerated. The efficacy in the treatment of pancreatic cancer was encouraging, but further studies would be required to prove its merit, compared with conventional treatment.
Collapse
Affiliation(s)
- Meng He
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Jinghua Sun
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Danyi Zhao
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Hongmei He
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Bing Wang
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Lingling Xu
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Yu Shang
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Shanling Ren
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Yang Zhang
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| | - Tao Wu
- a Department of Oncology , Second Affiliated Hospital of Dalian Medical University , Dalian , PR China
| |
Collapse
|
26
|
Lee C, Lim K, Kim SS, Thien LX, Lee ES, Oh KT, Choi HG, Youn YS. Chlorella-gold nanorods hydrogels generating photosynthesis-derived oxygen and mild heat for the treatment of hypoxic breast cancer. J Control Release 2019; 294:77-90. [PMID: 30543822 DOI: 10.1016/j.jconrel.2018.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/24/2018] [Accepted: 12/08/2018] [Indexed: 12/15/2022]
Abstract
Hypoxic tumors are rarely cured because their low oxygen environment restricts the cytotoxicity of many chemotherapeutics by blocking the production of intracellular reactive oxygen species. Inspired by the highly efficient production of oxygen as a waste product of chlorella photosynthesis, we developed an in situ rapidly gelling BSA-PEG-based hydrogel depot system that contains chlorella and gold nanorods, namely Chlorella AuNRs BSA-Gel. The entrapped chlorella in our gel efficiently generated a high level of oxygen and oxygenated hemoglobin via photosynthesis in response to 660-nm light in vitro and in vivo, respectively. The incorporated gold nanorods showed excellent performance in precisely increasing the surrounding temperature to 41-42 °C responding to 808-nm near-infrared laser, which presumably played a supporting role in expanding the tumor vasculature and thereby facilitating the delivery of doxorubicin and oxygen to hypoxic tumors of mice. Our results showed that a combined therapy of Chlorella AuNRs BSA-Gel plus Dox followed by irradiation at 660 nm and 808 nm significantly abolished 4 T1 breast cancer cell-xenografted tumors of BALB/C mice. We believe that our oxygen-generating and mild heat-emitting hydrogel should be considered to be a valid prototype of a local depot system for treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Changkyu Lee
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Kyungseop Lim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Sung Soo Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Le Xuan Thien
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eun Seong Lee
- Division of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan 15588, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
27
|
Chen J, Ning C, Zhou Z, Yu P, Zhu Y, Tan G, Mao C. Nanomaterials as photothermal therapeutic agents. PROGRESS IN MATERIALS SCIENCE 2019; 99:1-26. [PMID: 30568319 PMCID: PMC6295417 DOI: 10.1016/j.pmatsci.2018.07.005] [Citation(s) in RCA: 416] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Curing cancer has been one of the greatest conundrums in the modern medical field. To reduce side-effects associated with the traditional cancer therapy such as radiotherapy and chemotherapy, photothermal therapy (PTT) has been recognized as one of the most promising treatments for cancer over recent years. PTT relies on ablation agents such as nanomaterials with a photothermal effect, for converting light into heat. In this way, elevated temperature could kill cancer cells while avoiding significant side effects on normal cells. This theory works because normal cells have a higher heat tolerance than cancer cells. Thus, nanomaterials with photothermal effects have attracted enormous attention due to their selectivity and non-invasive attributes. This review article summarizes the current status of employing nanomaterials with photothermal effects for anti-cancer treatment. Mechanisms of the photothermal effect and various factors affecting photothermal performance will be discussed. Efficient and selective PTT is believed to play an increasingly prominent role in cancer treatment. Moreover, merging PTT with other methods of cancer therapies is also discussed as a future trend.
Collapse
Affiliation(s)
- Junqi Chen
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Chengyun Ning
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Zhengnan Zhou
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Peng Yu
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Oklahoma, United States
| | - Guoxin Tan
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Oklahoma, United States
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
28
|
Liao S, Hu X, Liu Z, Lin Y, Liang R, Zhang Y, Li Q, Li Y, Liao X. Synergistic action of microwave-induced mild hyperthermia and paclitaxel in inducing apoptosis in the human breast cancer cell line MCF-7. Oncol Lett 2019; 17:603-615. [PMID: 30655807 PMCID: PMC6313200 DOI: 10.3892/ol.2018.9629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 10/15/2018] [Indexed: 11/05/2022] Open
Abstract
Microwave mild hyperthermia and paclitaxel have been reported to be involved in variety of solid tumors. However, rare related researches have been accomplished via directly killing tumor cells using thermochemotherapy. In order to clarify the potential synergy between microwave-induced hyperthermia at temperatures <41°C and paclitaxel chemotherapy for inhibiting the growth of the human breast cancer cell line MCF-7, an MTT assay was used. The MCF-7 cells cultured in vitro were treated with paclitaxel alone, treated with microwave-induced hyperthermia for 2 h alone (at 40, 40.5 or 41°C), or treated with a combination of paclitaxel and 2 h of hyperthermia (at 40, 40.5 or 41°C). Flow cytometry was used to determine the cell apoptosis rate and it was demonstrated that paclitaxel decreased cell viability in a dose-dependent manner. Alone, hyperthermia for 2 h at 41°C induced apoptosis in MCF-7 cells, to a greater extent compared with hyperthermia for 2 h at 40.0 or 40.5°C (P<0.05). Together, paclitaxel and 2 h of hyperthermia at 40.5°C induced significantly increased apoptosis compared with either treatment alone (P<0.05). Increasing the temperature to 41°C in combination with paclitaxel increased the apoptotic ratio from 12.21±1.02% to 16.36±2.39%. The apoptotic ratio correlated positively with hyperthermia temperature and duration following hyperthermia, as did the synergistic effect obtained by combining hyperthermia and paclitaxel. Notably, the combination of 5 µg/ml paclitaxel and 2 h of hyperthermia at 40°C enhanced MCF-7 cell proliferation. Mild hyperthermia may exert anti-tumor effects by inducing apoptosis, and combining hyperthermia with paclitaxel synergistically induces apoptosis. Paclitaxel dose and hyperthermia temperature require careful optimization, as low-dose paclitaxel combined with hyperthermia at an insufficient temperature may enhance breast cancer proliferation.
Collapse
Affiliation(s)
- Sina Liao
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaohua Hu
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhihui Liu
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yan Lin
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rong Liang
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yumei Zhang
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qian Li
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yongqiang Li
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoli Liao
- Department of The First Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
29
|
Regional hyperthermia combined with radiotherapy for esophageal squamous cell carcinoma with supraclavicular lymph node metastasis. Oncotarget 2018; 8:5339-5348. [PMID: 28029663 PMCID: PMC5354912 DOI: 10.18632/oncotarget.14148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022] Open
Abstract
To assess the efficacy and toxicity of Intensity-modulated radiotherapy (IMRT) and hyperthermia for upper and middle thoracic esophageal squamous cell carcinoma (UMT-ESCC) with supraclavicular lymph node metastasis. A total of 50 patients with UMT-ESCC with supraclavicular lymph node metastasis were evaluated in this retrospective study. All patients received IMRT. Hyperthermia was delivered simultaneously with irradiation, in 45 minutes twice a week for 5-6 weeks. Hyperthermia included supraclavicular lymph node metastasis. Forty-four patients (88.0%) received concurrent chemoradiotherapy based on cisplatin regimens. The most common types of hematological toxicities were anemia (62.0%) and leukopenia (60.0%). Most of these events were grade 1-2 and transient. The 3-year progression-free survival (PFS) rate and overall survival (OS) rate were 34.9% and 42.5%, respectively. Cox regression revealed that tumor length and number of supraclavicular lymph node metastasis were two independent predictors of OS (tumor length: HR=3.65, p=0.008; nodal stage: HR=8.07, p=0.019). The IMRT combined with supraclavicular regional hyperthermia has low toxicity and well tolerated with excellent local control in UMT-ESCC with supraclavicular lymph node metastasis.
Collapse
|
30
|
Kargar S, Khoei S, Khoee S, Shirvalilou S, Mahdavi SR. Evaluation of the combined effect of NIR laser and ionizing radiation on cellular damages induced by IUdR-loaded PLGA-coated Nano-graphene oxide. Photodiagnosis Photodyn Ther 2017; 21:91-97. [PMID: 29155336 DOI: 10.1016/j.pdpdt.2017.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/04/2017] [Accepted: 11/15/2017] [Indexed: 12/23/2022]
Abstract
Glioma is one of the most common malignant cancers of the central nervous system (CNS). Radiatherapy and chemotherapy may be used to slow the growth of tumors that cannot be removed with surgery. The current study developed a combination therapy tool using Nanographene oxide (NGO) functionalized with poly lactic-co-glycolic acid (PLGA) as a carrier of 5-iodo-2-deoxyuridine (IUdR) for glioma cancer treatment. U87MG cells were treated in different groups with IUdR, PLGA-coated Nanographene oxide (PLGA-NGO), IUdR-loaded PLGA-coated Nanographene oxide (IUdR-PLGA-NGO), 2Gy 6MV X-ray radiation, and near-infrared region (NIR) laser radiation. PLGA-NGO showed excellent biocompatibility, high storage capacity for IUdR and high photothermal conversion efficiency. It was effectively employed to create cell damage in the U87MG cell line in the presence of X-ray (6 MV) and NIR laser. Moreover, IUdR-PLGA-NGO+X-ray+NIR laser significantly reduced the plating efficiency of the cells in comparison with IUdR-PLGA-NGO+X-ray and IUdR-PLGA-NGO+NIR laser. Furthermore, Prussian blue staining showed that IUdR-PLGA-NGO-SPIONs were delivered into glioblastoma cells. The PLGA-NGO loaded with IUdR under NIR and X-ray radiation exhibited the highest cytotoxicity toward U87MG cells when compared with other treatment methods, indicating efficient radio-photothermal targeted therapy.
Collapse
Affiliation(s)
- Samira Kargar
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samideh Khoei
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Khoee
- Department of Polymer Chemistry, School of Sciences, University of Tehran, Tehran, Iran
| | - Sakine Shirvalilou
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seied Rabi Mahdavi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Ojha T, Pathak V, Shi Y, Hennink WE, Moonen CTW, Storm G, Kiessling F, Lammers T. Pharmacological and physical vessel modulation strategies to improve EPR-mediated drug targeting to tumors. Adv Drug Deliv Rev 2017; 119:44-60. [PMID: 28697952 PMCID: PMC5919100 DOI: 10.1016/j.addr.2017.07.007] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 06/22/2017] [Accepted: 07/06/2017] [Indexed: 02/08/2023]
Abstract
The performance of nanomedicine formulations depends on the Enhanced Permeability and Retention (EPR) effect. Prototypic nanomedicine-based drug delivery systems, such as liposomes, polymers and micelles, aim to exploit the EPR effect to accumulate at pathological sites, to thereby improve the balance between drug efficacy and toxicity. Thus far, however, tumor-targeted nanomedicines have not yet managed to achieve convincing therapeutic results, at least not in large cohorts of patients. This is likely mostly due to high inter- and intra-patient heterogeneity in EPR. Besides developing (imaging) biomarkers to monitor and predict EPR, another strategy to address this heterogeneity is the establishment of vessel modulation strategies to homogenize and improve EPR. Over the years, several pharmacological and physical co-treatments have been evaluated to improve EPR-mediated tumor targeting. These include pharmacological strategies, such as vessel permeabilization, normalization, disruption and promotion, as well as physical EPR enhancement via hyperthermia, radiotherapy, sonoporation and phototherapy. In the present manuscript, we summarize exemplary studies showing that pharmacological and physical vessel modulation strategies can be used to improve tumor-targeted drug delivery, and we discuss how these advanced combination regimens can be optimally employed to enhance the (pre-) clinical performance of tumor-targeted nanomedicines.
Collapse
Affiliation(s)
- Tarun Ojha
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Vertika Pathak
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany
| | - Yang Shi
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Chrit T W Moonen
- Imaging division, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG, Utrecht, The Netherlands; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Fabian Kiessling
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany.
| | - Twan Lammers
- Department of Nanomedicines and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic, 52074 Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG, Utrecht, The Netherlands; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands.
| |
Collapse
|
32
|
Hornung M, Werner JM, Schlitt HJ. Applications of hyperthermic intraperitoneal chemotherapy for metastatic colorectal cancer. Expert Rev Anticancer Ther 2017; 17:841-850. [PMID: 28715968 DOI: 10.1080/14737140.2017.1357470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) plays a pivotal role in the current treatment of peritoneal carcinomatosis (PC) from colorectal cancer (CRC). Since the first demonstration, benefits for patients and especially an increase in survival have been described. In recent years, feasibility, efficacy and safety of HIPEC have been improved and progress has been made in understanding its oncological mechanism. Areas covered: In this article, leading publications have been reviewed including clinical trials to describe the clinical presentation of PC due to CRC and present recent evidence of the CRS/HIPEC procedure. The surgical approach including evaluation of the extent of PC is described and, in addition, the article reports about different HIPEC techniques as well as several protocols. Furthermore, the development and prognostic benefit of the combination of intraperitoneal and intravenous chemotherapy are outlined. Consideration has been given in particular to patient selection and the use of HIPEC if complete cytoreduction is not feasible. Expert commentary: The CRS/HIPEC procedure represents a curative approach to treat patients with PC from CRC. However, surgical skills and the HIPEC technique still require specialized oncological centers.
Collapse
Affiliation(s)
- Matthias Hornung
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| | - Jens M Werner
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| | - Hans J Schlitt
- a Department of Surgery , University of Regensburg , Regensburg , Germany
| |
Collapse
|
33
|
Mohammadi MR, Nojoomi A, Mozafari M, Dubnika A, Inayathullah M, Rajadas J. Nanomaterials engineering for drug delivery: a hybridization approach. J Mater Chem B 2017; 5:3995-4018. [PMID: 32264132 DOI: 10.1039/c6tb03247h] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The last twenty years have witnessed great advances in biology, medicine, and materials science, leading to the development of various nanoparticle (NP)-mediated drug delivery systems. Innovation in materials science has led the generation of biodegradable, biocompatible, stimuli-responsive, and targeted delivery systems. However, currently available nanotherapeutic technologies are not efficient, which has culminated in the failure of their clinical trials. Despite huge efforts devoted to drug delivery nanotherapeutics, only a small amount of the injected material could reach the desired target. One promising strategy to enhance the efficiency of NP drug delivery is to hybridize multiple materials, where each component could play a critical role in an efficient multipurpose delivery system. This review aims to comprehensively cover different techniques, materials, advantages, and drawbacks of various systems to develop hybrid nano-vesicles for drug delivery. Attention is finally given to the hybridization benefits in overcoming the biological barriers for drug delivery. It is believed that the advent of modern nano-formulations for multifunctional hybrid carriers paves the way for future advances to achieve more efficient drug delivery systems.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, 1050 Arastradero Road, Palo Alto, CA 94304, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ranjan A, Benjamin CJ, Negussie AH, Chokshi S, Chung PH, Volkin D, Yeram N, Linehan WM, Dreher MR, Pinto PA, Wood BJ. Biodistribution and Efficacy of Low Temperature-Sensitive Liposome Encapsulated Docetaxel Combined with Mild Hyperthermia in a Mouse Model of Prostate Cancer. Pharm Res 2016; 33:2459-69. [PMID: 27343000 PMCID: PMC7641880 DOI: 10.1007/s11095-016-1971-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/09/2016] [Indexed: 01/21/2023]
Abstract
PURPOSE Low temperature sensitive liposome (LTSL) encapsulated docetaxel were combined with mild hyperthermia (40-42°C) to investigate in vivo biodistribution and efficacy against a castrate resistant prostate cancer. METHOD Female athymic nude mice with human prostate PC-3 M-luciferase cells grown subcutaneously into the right hind leg were randomized into six groups: saline (+/- heat), free docetaxel (+/- heat), and LTSL docetaxel (+/- heat). Treatment (15 mg docetaxel/kg) was administered via tail vein once tumors reached a size of 200-300 mm(3). Mice tumor volumes and body weights were recorded for up to 60 days. Docetaxel concentrations of harvested tumor and organ/tissue homogenates were determined by LC-MS. Histological evaluation (Mean vessel density, Ki67 proliferation, Caspase-3 apoptosis) of saline, free Docetaxel and LTSL docetaxel (+/- heat n = 3-5) was performed to determine molecular mechanism responsible for tumor cell killing. RESULT LTSL/heat resulted in significantly higher tumor docetaxel concentrations (4.7-fold greater compared to free docetaxel). Adding heat to LTSL Docetaxel or free docetaxel treatment resulted in significantly greater survival and growth delay compared to other treatments (p < 0.05). Differences in body weight between all Docetaxel treatments were not reduced by >10% and were not statistically different from each other. Molecular markers such as caspase-3 were upregulated, and Ki67 expression was significantly decreased in the chemo-hyperthermia group. Vessel density was similar post treatment, but the heated group had reduced vessel area, suggesting thermal enhancement in efficacy by reduction in functional perfusion. CONCLUSION This technique of hyperthermia sensitization and enhanced docetaxel delivery has potential for clinical translation for prostate cancer treatment.
Collapse
Affiliation(s)
- Ashish Ranjan
- Center for Interventional Oncology, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, MSC 1182- building 10- room 1c -341, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Compton J Benjamin
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ayele H Negussie
- Center for Interventional Oncology, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, MSC 1182- building 10- room 1c -341, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Saurin Chokshi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Paul H Chung
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dmitry Volkin
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nitin Yeram
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew R Dreher
- Center for Interventional Oncology, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, MSC 1182- building 10- room 1c -341, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, MSC 1182- building 10- room 1c -341, 10 Center Drive, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
35
|
Abramian A, Zivanovic O, Kuhn W, Weber S, Schaefer N, Keyver-Paik MD, Kiefer N. Introducing Hyperthermic Intraperitoneal Chemotherapy into Gynecological Oncology Practice - Feasibility and Safety Considerations: Single-Center Experience. Oncol Res Treat 2016; 39:178-84. [PMID: 27160457 DOI: 10.1159/000445180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/03/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Within the surgical oncology community interest is increasingly focusing on combining surgical cytoreduction and regional chemotherapeutic drug delivery to manage solid abdominal tumors. In particular, the role of hyperthermic intraperitoneal chemotherapy (HIPEC) is evolving for treating epithelial ovarian carcinomas (EOCs), as EOCs remain confined to the peritoneal cavity for most of their natural history. Currently there is no evidence from prospective trials to confirm an overall survival benefit associated with HIPEC. In addition, there are no generally accepted regimens, which results in heterogeneous clinical procedures. METHODS We have initiated a HIPEC program at our institution and completed a phase I study of HIPEC with cisplatin in patients with platinum-sensitive recurrent EOC. The data have been published and prove the feasibility of this approach. In the process of introducing HIPEC, several safety measures had to be taken into consideration. RESULTS We present the implications and requirements of introducing HIPEC in clinical practice and discuss our proposed procedure referring to the recent literature. CONCLUSION HIPEC is feasible and can be performed safely in daily gynecological oncology routine provided that certain considerations and precautions are taken into account during its introduction to guarantee a proper and safe operating sequence.
Collapse
Affiliation(s)
- Alina Abramian
- Department of Obstetrics and Gynecology, Center for Integrated Oncology, University Hospital Bonn, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
van der Heijden AG, Dewhirst MW. Effects of hyperthermia in neutralising mechanisms of drug resistance in non-muscle-invasive bladder cancer. Int J Hyperthermia 2016; 32:434-45. [DOI: 10.3109/02656736.2016.1155761] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
37
|
Lungoci C, Mironiuc AI, Muntean V, Oniu T, Leebmann H, Mayr M, Piso P. Multimodality treatment strategies have changed prognosis of peritoneal metastases. World J Gastrointest Oncol 2016; 8:67-82. [PMID: 26798438 PMCID: PMC4714147 DOI: 10.4251/wjgo.v8.i1.67] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/22/2015] [Accepted: 11/11/2015] [Indexed: 02/05/2023] Open
Abstract
For a long time, treatment of peritoneal metastases (PM) was mostly palliative and thus, this status was link with “terminal status/despair”. The current multimodal treatment strategy, consisting of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), has been strenuously achieved over time, but seems to be the best treatment option for PM patients. As we reviewed the literature data, we could emphasize some milestones and also, controversies in the history of proposed multimodal treatment and thus, outline the philosophy of this approach, which seems to be an unusual one indeed. Initially marked by nihilism and fear, but benefiting from a remarkable joint effort of human and material resources (multi-center and -institutional research), over a period of 30 years, CRS and HIPEC found their place in the treatment of PM. The next 4 years were dedicated to the refinement of the multimodal treatment, by launching research pathways. In selected patients, with requires training, it demonstrated a significant survival results (similar to the Hepatic Metastases treatment), with acceptable risks and costs. The main debates regarding CRS and HIPEC treatment were based on the oncologists’ perspective and the small number of randomized clinical trials. It is important to statement the PM patient has the right to be informed of the existence of CRS and HIPEC, as a real treatment resource, the decision being made by multidisciplinary teams.
Collapse
|
38
|
Li X, Yang Z, Hu N, Zhang L, Zhang Y, Yin L. Docetaxel-loaded SiO2@Au@GO core–shell nanoparticles for chemo-photothermal therapy of cancer cells. RSC Adv 2016. [DOI: 10.1039/c6ra03886g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The multifunctional antitumor SiO2@Au@GO core–shell nanoparticles (NPs) have been fabricated for serving as drug nanocarrier and photothermal inducer to have a treatment on human prostate cancer cell DU 145.
Collapse
Affiliation(s)
- Xiaolin Li
- Department of Urology
- Changzheng Hospital
- The Second Military Medical University
- Shanghai 200003
- PR China
| | - Zhi Yang
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education
- Department of Micro/Nano Electronics
- School of Electronic Information and Electrical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Nantao Hu
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education
- Department of Micro/Nano Electronics
- School of Electronic Information and Electrical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Liying Zhang
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education
- Department of Micro/Nano Electronics
- School of Electronic Information and Electrical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Yafei Zhang
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education
- Department of Micro/Nano Electronics
- School of Electronic Information and Electrical Engineering
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Lei Yin
- Department of Urology
- Changzheng Hospital
- The Second Military Medical University
- Shanghai 200003
- PR China
| |
Collapse
|
39
|
Lee SY, Kim MG. The effect of modulated electro-hyperthermia on the pharmacokinetic properties of nefopam in healthy volunteers: A randomised, single-dose, crossover open-label study. Int J Hyperthermia 2015; 31:869-74. [PMID: 26507458 PMCID: PMC4776683 DOI: 10.3109/02656736.2015.1095358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Purpose: Nefopam is a widely available analgesic for the management of pain. The aim of this study was to reveal the effect of regional hyperthermia of the abdominal area on the pharmacokinetics of nefopam. Materials and methods: A randomised, single-dose, crossover, open-label study was conducted to reveal the effect of hyperthermia using modulated electro-hyperthermia on the pharmacokinetics of nefopam. The pharmacokinetics of orally administered nefopam without hyperthermia was studied in 12 healthy volunteers and then 7 days later they were treated with nefopam plus modulated electro-hyperthermia to the abdominal area for 1 h. Blood samples were collected up to 24 h after the drug administration. From the blood concentration-time curve, the maxinum plasma concentration (Cmax), time to Cmax (Tmax) and the area under the curve (AUC) were obtained. The safety and tolerability of these treatments were also assessed. Results: The geometric mean ratios (GMRs) ((nefopam + modulated electro-hyperthermia)/nefopam) and the associated 90% confidence intervals (CIs) for Cmax, AUClast and AUCinf were 1.2804 (1.1155∼1.4696), 1.0512 (0.9555∼1.1566) and 1.0612 (0.9528∼1.1819), respectively. The increase in Cmax was statistically significant, and Tmax was significantly shortened. Conclusions: The significant increase in Cmax and decrease in Tmax indicated that modulated electro-hyperthermia increased the absorption of the orally administered nefopam, thereby transitionally increasing the blood concentration of the drug. The AUC is an important parameter that contributes to the therapeutic effect of drugs. The lack of significant change in AUC suggests that modulated electro-hyperthermia may increases the absorption of orally administered drugs without increasing the systemic adverse effect of the drugs.
Collapse
Affiliation(s)
- Sun Young Lee
- a Department of Radiation Oncology , Chonbuk National University Hospital , Jeonju , Jeonbuk , Republic of Korea .,c Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute, Chonbuk National University Hospital , Jeonju , Jeonbuk , Republic of Korea
| | - Min-Gul Kim
- b Clinical Pharmacology Unit and Biomedical Research Institute, Chonbuk National University Hospital , Jeonju , Jeonbuk , Republic of Korea , and.,c Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute, Chonbuk National University Hospital , Jeonju , Jeonbuk , Republic of Korea
| |
Collapse
|
40
|
Helm CW. Hyperthermic intraperitoneal chemotherapy for ovarian cancer: is there a role? J Gynecol Oncol 2015; 26:1-2. [PMID: 25609161 PMCID: PMC4302278 DOI: 10.3802/jgo.2015.26.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
41
|
Oei AL, Vriend LEM, Crezee J, Franken NAP, Krawczyk PM. Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol 2015; 10:165. [PMID: 26245485 PMCID: PMC4554295 DOI: 10.1186/s13014-015-0462-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/13/2015] [Indexed: 12/03/2022] Open
Abstract
The currently available arsenal of anticancer modalities includes many DNA damaging agents that can kill malignant cells. However, efficient DNA repair mechanisms protect both healthy and cancer cells against the effects of treatment and contribute to the development of drug resistance. Therefore, anti-cancer treatments based on inflicting DNA damage can benefit from inhibition of DNA repair. Hyperthermia – treatment at elevated temperature – considerably affects DNA repair, among other cellular processes, and can thus sensitize (cancer) cells to DNA damaging agents. This effect has been known and clinically applied for many decades, but how heat inhibits DNA repair and which pathways are targeted has not been fully elucidated. In this review we attempt to summarize the known effects of hyperthermia on DNA repair pathways relevant in clinical treatment of cancer. Furthermore, we outline the relationships between the effects of heat on DNA repair and sensitization of cells to various DNA damaging agents.
Collapse
Affiliation(s)
- Arlene L Oei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands. .,Department of Radiotherapy, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Lianne E M Vriend
- Van Leeuwenhoek Centre for Advanced Microscopy (LCAM)-AMC, Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| | - Johannes Crezee
- Department of Radiotherapy, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Nicolaas A P Franken
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands. .,Department of Radiotherapy, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| | - Przemek M Krawczyk
- Van Leeuwenhoek Centre for Advanced Microscopy (LCAM)-AMC, Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
42
|
Yonemura Y, Canbay E, Endou Y, Ishibashi H, Mizumoto A, Li Y, Liu Y, Takeshita K, Ichinose M, Takao N, Saitou T, Noguchi K, Hirano M, Glehen O, Brűcher B, Sugarbaker PH. Comprehensive treatment for the peritoneal metastasis from gastric cancer. World J Surg Proced 2015; 5:187-197. [DOI: 10.5412/wjsp.v5.i2.187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/05/2014] [Accepted: 03/18/2015] [Indexed: 02/06/2023] Open
Abstract
Recently, a novel comprehensive treatment consisting of cytoreductive surgery (CRS) and perioperative chemotherapy (POC) was developed for the treatment of peritoneal metastasis (PM) with a curative intent. In the treatment, the macroscopic disease is completely removed by the peritonectomy techniques in combination with POC. This article reviews the results of the comprehensive treatment for PM from gastric cancer, and verifies the effects of CRS and POC, including neoadjuvant chemotherapy (NAC) and hyperthermic intraoperative intraperitoneal chemotherapy (HIPEC). Completeness of cytoreduction, peritoneal carcinomatosis index (PCI) less than the threshold levels after NAC, absence of ascites, cytologic status, pathologic response after NAC are the independent prognostic factors. Among these prognostic factors, PCI threshold level is the most valuable independent prognostic factor. After staging laparoscopy, patients with PM from gastric cancer are recommended to treat with NAC before CRS. After NAC, indication for CRS is determined by laparoscopy. The indications of the comprehensive treatment are patients with PCI less than the threshold levels, negative cytology, and responders after NAC. Patients satisfy these factors are the candidates for the CRS and HIPEC.
Collapse
|
43
|
To KKW, Poon DC, Wei Y, Wang F, Lin G, Fu L. Pelitinib (EKB-569) targets the up-regulation of ABCB1 and ABCG2 induced by hyperthermia to eradicate lung cancer. Br J Pharmacol 2015; 172:4089-106. [PMID: 25988710 DOI: 10.1111/bph.13189] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/15/2015] [Accepted: 05/07/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Pelitinib is a potent irreversible EGFR TK inhibitor currently in clinical trials for the treatment of lung cancer. Hyperthermia has been applied concomitantly with chemotherapy and radiotherapy to enhance treatment outcome. In this study, we investigated the ability of the combination of pelitinib with other conventional anticancer drugs to specifically target cancer cells with up-regulated efflux transporters ABCB1/ABCG2 after hyperthermia as a novel way to eradicate the cancer stem-like cells responsible for cancer recurrence. EXPERIMENTAL APPROACH Alterations in intracellular topotecan accumulation, the efflux of fluorescent probe substrates, expression and ATPase activity of ABCB1/ABCG2 and tumoursphere formation capacity of side population (SP) cells sorted after hyperthermia were examined to elucidate the mechanism of pelitinib-induced chemosensitization. KEY RESULTS While pelitinib did not modulate ABCB1/ABCG2 expressions, the combination of pelitinib with transporter substrate anticancer drugs induced more marked apoptosis, specifically in cells exposed to hyperthermia. The flow cytometric assay showed that both ABCB1- and ABCG2-mediated drug effluxes were significantly inhibited by pelitinib in a concentration-dependent manner. The inhibition kinetics suggested that pelitinib is a competitive inhibitor of ABCB1/ABCG2, which is consistent with its ability to stimulate their ATPase activity. SP cells sorted after hyperthermia were found to be more resistant to anticancer drugs, presumably due to the up-regulation of ABCB1 and ABCG2. Importantly, pelitinib specifically enhanced the chemosensitivity but reduced the tumoursphere formation capacity of these SP cells. CONCLUSIONS AND IMPLICATIONS This study demonstrated a novel approach, exploiting drug resistance, to selectively kill cancer stem-like cells after hyperthermia.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Daniel C Poon
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuming Wei
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
44
|
Braam HJ, Schellens JH, Boot H, van Sandick JW, Knibbe CA, Boerma D, van Ramshorst B. Selection of chemotherapy for hyperthermic intraperitoneal use in gastric cancer. Crit Rev Oncol Hematol 2015; 95:282-96. [PMID: 25921419 DOI: 10.1016/j.critrevonc.2015.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/22/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Several studies have shown the potential benefit of cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) in gastric cancer patients. At present the most effective chemotherapeutic regime in HIPEC for gastric cancer is unknown. The aim of this review was to provide a comprehensive overview of chemotherapeutic agents used for HIPEC in gastric cancer. METHODS A literature search was conducted using the PubMed database to identify studies on chemotherapy used for HIPEC in gastric cancer patients. RESULTS AND CONCLUSION The chemotherapeutic regime of choice in HIPEC for gastric cancer has yet to be determined. The wide variety in studies and study parameters, such as chemotherapeutic agents, dosage, patient characteristics, temperature of perfusate, duration of perfusion, carrier solutions, intraperitoneal pressure and open or closed perfusion techniques, warrant more experimental and clinical studies to determine the optimal treatment schedule. A combination of drugs probably results in a more effective treatment.
Collapse
Affiliation(s)
- H J Braam
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands.
| | - J H Schellens
- Division of Clinical Pharmacology, Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Science Faculty, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - H Boot
- Division of Gastroenterology and Hepatology, Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J W van Sandick
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C A Knibbe
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - D Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - B van Ramshorst
- Department of Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
45
|
Movahedi F, Hu RG, Becker DL, Xu C. Stimuli-responsive liposomes for the delivery of nucleic acid therapeutics. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1575-84. [PMID: 25819885 DOI: 10.1016/j.nano.2015.03.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/07/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
UNLABELLED Nucleic acid therapeutics (NATs) are valuable tools in the modulation of gene expression in a highly specific manner. So far, NATs have been actively pursued in both pre-clinical and clinical studies to treat diseases such as cancer, infectious and inflammatory diseases. However, the clinical application of NATs remains a considerable challenge owing to their limited cellular uptake, low biological stability, off-target effect, and unfavorable pharmacokinetics. One concept to address these issues is to deliver NATs within stimuli-responsive liposomes, which release their contents of NATs upon encountering environmental changes such as temperature, pH, and ion strength. In this case, before reaching the targeted tissue/organ, NATs are protected from degradation by enzymes and immune system. Once at the area of interest, localized and targeted delivery can be achieved with minimal influence to other parts of the body. Here, we discuss the latest developments and existing challenges in this field. FROM THE CLINICAL EDITOR Nucleic acid therapeutics have been shown to enhance or eliminate specific gene expression in experimental research. Unfortunately, clinical applications have so far not been realized due to problems of easy degradation and possible toxicity. The use of nanosized carriers such as liposomes to deliver nucleic acids is one solution to overcome these problems. In this review article the authors describe and discuss the potentials of various trigger-responsive "smart" liposomes, with a view to help other researchers to design better liposomal nucleic acid delivery systems.
Collapse
Affiliation(s)
- Fatemeh Movahedi
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Rebecca G Hu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore.
| |
Collapse
|
46
|
Yonemura Y, Canbay E, Endou Y, Ishibashi H, Mizumoto A, Miura M, Li Y, Liu Y, Takeshita K, Ichinose M, Takao N, Hirano M, Sako S, Tsukiyama G. Peritoneal cancer treatment. Expert Opin Pharmacother 2014; 15:623-36. [PMID: 24617975 DOI: 10.1517/14656566.2014.879571] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION In the past, peritoneal surface malignancy (PSM) was considered as a final stage of cancer, and patients were offered the best supportive care. Recently, a new therapeutic alternative approach based on the combination of surgery with chemotherapy was developed. In this curative intent, the macroscopic disease was treated with cytoreductive surgery (CRS) combined with perioperative chemotherapy, including neoadjuvant chemotherapy, hyperthermic intraoperative intraperitoneal chemotherapy, extensive intraoperative peritoneal lavage and early postoperative intraperitoneal chemotherapy AREAS COVERED This article reviews the mechanisms of the formation of PSM, quantitative estimation of PSM and residual disease, multimodal treatment, value of laparoscopy, prognostic factors and patients' selection for the multimodal therapy. EXPERT OPINION Recent studies show that CRS plus intraperitoneal chemotherapy applications confer prolonged survival in patients with PSM from colorectal, gastric, ovarian, appendiceal mucinous carcinoma and diffuse malignant peritoneal mesothelioma. The comprehensive treatment is now justified as state-of-the-art for patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Yutaka Yonemura
- NPO Organization to Support Peritoneal Surface Malignancy Treatment , Oosaka , Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Al-Badawi IA, Abu-Zaid A, Azzam A, AlOmar O, AlHusaini H, Amin T. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for management of recurrent/relapsed ovarian granulosa cell tumor: a single-center experience. J Obstet Gynaecol Res 2014; 40:2066-2075. [PMID: 25181627 DOI: 10.1111/jog.12460] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/31/2014] [Indexed: 02/05/2023]
Abstract
AIM The aim of this study was to retrospectively report our experience (efficacy/morbidity) with cytoreductive surgery+hyperthermic intraperitoneal chemotherapy (CRS+HIPEC) for the management of recurrent/relapsed ovarian granulosa cell tumors (OGCT). MATERIAL AND METHODS From 2010 to 2013, six patients underwent CRS+HIPEC. CRS was performed with standard peritonectomy procedures and visceral resections directed towards complete elimination of tumors from the abdominopelvic cavity. HIPEC was performed with cisplatin (50 mg/m²) and doxorubicin (15 mg/m²) and allowed to circulate in the abdominopelvic cavity for 90 min at 41.0-42.2°C. RESULTS Cytoreduction completeness (CC-0) was achieved in all except one patient (CC-1). Five patients had OGCT recurrences in abdomen+pelvis and one patient in abdomen only. No grade V morbidity (Clavien-Dindo classification) occurred. Two patients developed lung atelectasis, which was managed by mere chest physiotherapy (grade I). One patient developed urinary tract infection (grade II) and another patient developed pneumonia (grade II) - both of which were managed by antibiotics. One patient developed splenic bed and anterior abdominal wall collections requiring ultrasound-guided aspiration without general anesthesia (grade III). One patient developed pulmonary embolism requiring intensive care-unit management (grade IV). Four chemo-naïve patients received adjuvant chemotherapy whereas the remaining two previously chemo-exposed patients received no adjuvant therapy. All patients were alive and disease-free without proof of recurrence/relapse at 40, 32, 27, 24, 20 and 16 months. The average interval of follow-up after CRS+HIPEC was roughly 27 months (range: 16-40 months). CONCLUSION CRS+HIPEC appears to be an efficacious and morbidly well-tolerated therapeutic modality for recurrent/relapsed OGCT. Long-term follow-up data and further research are needed.
Collapse
Affiliation(s)
- Ismail A Al-Badawi
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | | | | | | |
Collapse
|
48
|
Shirafuji A, Shinagawa A, Kurokawa T, Yoshida Y. Locally-advanced unresected uterine leiomyosarcoma with triple-modality treatment combining radiotherapy, chemotherapy and hyperthermia: A case report. Oncol Lett 2014; 8:637-641. [PMID: 25009648 PMCID: PMC4081420 DOI: 10.3892/ol.2014.2193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 05/07/2014] [Indexed: 11/05/2022] Open
Abstract
Advanced uterine leiomyosarcoma (LMS) is a rare and extremely aggressive disease. In patients with advanced and unresected uterine LMS, multidisciplinary therapy is the best treatment option, although no consensus exists on the efficacy of the treatment. The present study describes the case of a 41-year-old female who underwent laparotomy due to a large uterine tumor. Exploratory laparotomy revealed a large tumor that had extended from the pelvic wall to the outside of the pelvis and then invaded the colon. Large residual tumors remained present in the pelvis following suboptimal debulking surgery. Subsequent to surgery, the patient was treated with adjuvant radiotherapy, followed by chemotherapy with regional whole pelvis hyperthermia (HT). Computed tomography revealed stable disease prior and subsequent to combination treatment. While treatment was being administered for third/fourth-degree burns and subcutaneous fatty necrosis, the patient developed multi-organ failure and succumbed. The present case report describes the potential for using a combination of chemotherapy, HT and radiotherapy in patients with LMS. The development of an effective protocol is required for the administration of chemotherapy, HT and radiotherapy in patients with advanced unresected LMS.
Collapse
Affiliation(s)
- Aya Shirafuji
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan ; Department of Obstetrics and Gynecology, Fukui Aiiku Hospital, Fukui, Japan
| | - Akiko Shinagawa
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tetsuji Kurokawa
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
49
|
Abu-Zaid A, Azzam AZ, AlOmar O, Salem H, Amin T, Al-Badawi IA. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for managing peritoneal carcinomatosis from endometrial carcinoma: a single-center experience of 6 cases. Ann Saudi Med 2014; 34:159-166. [PMID: 24894786 PMCID: PMC6074854 DOI: 10.5144/0256-4947.2014.159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Endometrial carcinoma is the most common gynecologic malignancy worldwide. Prognosis of patients with peritoneal carcinomatosis (PC) from endometrial carcinoma is deadly, with an estimated median survival not exceeding 12 months. The objective of this study was to report our experience with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) for managing PC from primary and recurrent endometrial carcinoma. DESIGN AND SETTINGS A retrospective analysis of 6 patients with PC arising from endometrial cancer, who were managed with CRS and HIPEC at our referral tertiary care center, from November 2010 to August 2013. MATERIALS AND METHODS Six patients underwent CRS and HIPEC. CRS was performed using standard peritonectomy procedures and visceral resections directed toward the complete elimination of tumors from ab.dominopelvic cavity. HIPEC was performed with cisplatin (50 mg/m2) and doxorubicin (15 mg/m2) and allowed to circulate in abdominopelvic cavity for 90 minutes at 41.0 to 42.2°C. RESULTS Two patients with primary endometrial carcinoma and 4 patients with recurrent endometrial carcino.ma confined to peritoneal cavity were studied. Complete cytoreduction (CC-0) was achieved in 5 patients. The International Federation of Gynecology and Obstetrics (FIGO) stages and histopathological types were as follows: IB endometrioid adenocarcinomas (n=1), IC mesonephric carcinomas (n=1), IIIA endometrioid adenocarcino.mas (n=2), IIIA papillary serous carcinomas (n=1), and IIIC clear-cell carcinomas (n=1). Anastomotic leak (grade I) was the most commonly encountered postoperative complication. Two patients developed grade IV compli.cations due to septicemia and pulmonary embolism. No intraoperative mortality occurred. Postoperatively, all patients received chemotherapy (carboplatin and paclitaxel). In 1 patient, the clear-cell carcinoma histologic lesion relapsed within 6 months; the metastases spread to hepatic, pelvic, and mesenteric lymph nodes, and the patient died 5 months later. One patient with cytoreduction completeness of CC-2 developed hepatic metastases within 3 months and is still alive at a follow-up up 6 months. Remaining patients (n=4) are alive and disease free without evidence of recurrence of follow-ups at 35, 34, 19, and 7 months. CONCLUSION CRS and HIPEC are well-tolerated and feasibly promising management modalities in PC from primary and recurrent endometrial carcinoma. Further research is needed for in-depth analysis.
Collapse
Affiliation(s)
| | | | | | | | | | - Ismail A Al-Badawi
- Dr. Ismail A. Al-Badawi, MBC 52 Department of Obstetrics and Gynecology,, PO Box 3354, King Faisal Specialist Hospital and Research Centre,, Riyadh 11211, Saudi Arabia, T: +966-11- 442-7392, F: +966-11-442-7393,
| |
Collapse
|
50
|
Shanmugam V, Selvakumar S, Yeh CS. Near-infrared light-responsive nanomaterials in cancer therapeutics. Chem Soc Rev 2014; 43:6254-87. [DOI: 10.1039/c4cs00011k] [Citation(s) in RCA: 638] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Near-infrared light sensitive nanomaterials provide ideal nanoplatforms in site specific noninvasive cancer therapy.
Collapse
Affiliation(s)
| | - S. Selvakumar
- Department of Chemistry, Center for Micro/Nano Science and Technology
- and Advanced Optoelectronic Technology Center
- National Cheng Kung University
- Tainan 701, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, Center for Micro/Nano Science and Technology
- and Advanced Optoelectronic Technology Center
- National Cheng Kung University
- Tainan 701, Taiwan
| |
Collapse
|