Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 569
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Bull Cancer ; 111(3): 285-290, 2024 Mar.
Article in French | MEDLINE | ID: mdl-38331695

ABSTRACT

After more than a decade of good results using the combination of cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of peritoneal carcinosis of colorectal origin, the PRODIGE7 study, which specifically evaluated the role of HIPEC, failed to show any superiority in terms of overall and disease-free survival for the CRS+HIPEC combination compared with CRS alone. This study constituted a radical change in the knowledge and therapeutic attitudes observed to date. After reviewing the literature and the consensus of national and international experts, a synthesis is provided, together with an outlook on the questions raised and the therapeutic trials and innovations of the near future. An analysis of recent advances due to the advent of a new technique, PIPAC, is also proposed, as well as a review of current therapeutic trials in this field.


Subject(s)
Carcinoma , Colorectal Neoplasms , Hyperthermia, Induced , Peritoneal Neoplasms , Humans , Hyperthermic Intraperitoneal Chemotherapy , Hyperthermia, Induced/methods , Chemotherapy, Cancer, Regional Perfusion/methods , Colorectal Neoplasms/drug therapy , Carcinoma/therapy , Peritoneal Neoplasms/drug therapy , Combined Modality Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Survival Rate
3.
Int Immunopharmacol ; 111: 109166, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994852

ABSTRACT

INTRODUCTION: Heated intraperitoneal chemotherapy (HIPEC) is currently implemented in the treatment of peritoneal metastases from colorectal carcinoma (PM-CRC) origin. However, recurrence is common and the effectiveness of HIPEC has been questioned. The aim of this study was to evaluate the use of thymosin alpha 1 (Tα1), an immunomodulatory molecule, as an adjuvant to HIPEC treatment. METHODS: We developed an experimental model of HIPEC by the induction of PM-CRC in C57BL mice and intra-abdominal perfusion of mitomycin C (MMC). Mice were treated with Tα1 at 0.6 mg/kg for 5 days after HIPEC. Clinical and immunological parameters were compared between HIPEC and HIPEC + Tα1 groups. RESULTS: Treatment with Tα1 increased overall survival of mice compared to HIPEC treatment alone and sham-treated animals (16.1 ± 0.8 vs. 14.1 ± 0.6 and 11.8 ± 0.8, respectively, p = 0.02). Tα1 had no direct anti-tumor effect, as seen by lack of inhibition of tumor cell proliferation. Tα1 treatment induced a T helper (Th) 1 immune response in tumor metastases as evidenced by a significant increase of the Th1-specific markers IFN-γ and T-bet (1.21 ± 0.3 vs. 0.52 ± 0.08, p < 0.05; 0.88 ± 0.04 vs. 0.64 ± 0.14, p < 0.05, respectively). This Th1 skew was accompanied by increased CD8+ infiltration into omental and visceral metastases by Tα1 treatment compared to sham and HIPEC-treated animals (21.24 ± 2.16 vs. 10.45 ± 0.89 and 7.7 ± 1.3, p < 0.001; 14.12 ± 1.54 vs. 12.12 ± 0.01 and 6.64 ± 0.87, p < 0.01, respectively). CONCLUSIONS: Tα1 augments the effect of HIPEC by the induction of a Th1 anti-tumor immune response. Further experiments should evaluate Tα1 and other novel immunomodulators in order to exploit the immunological opportunities created by HIPEC.


Subject(s)
Carcinoma , Colonic Neoplasms , Colorectal Neoplasms , Hyperthermia, Induced , Peritoneal Neoplasms , Animals , Carcinoma/therapy , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Combined Modality Therapy , Hyperthermic Intraperitoneal Chemotherapy , Mice , Mice, Inbred C57BL , Models, Theoretical , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Thymalfasin/therapeutic use
4.
Rev Med Suisse ; 18(782): 990-996, 2022 May 18.
Article in French | MEDLINE | ID: mdl-35583278

ABSTRACT

Ovarian cancer is the first cause of death by gynecological cancer. Most of the patients are diagnosed with peritoneal carcinomatosis that represents a therapeutic challenge. Its management implies maximal cytoreductive surgery with survival benefit. Over the last three decades, several strategies of intra-peritoneal chemotherapy have been investigated. This includes intra-peritoneal adjuvant chemotherapy that is used mainly in North America, hyperthermic intraperitoneal chemotherapy (HIPEC) and more recently pressurized intraperitoneal aerosol chemotherapy (PIPAC). In the current article, we review the evidence in favor of each therapeutic approach, and we propose treatment algorithms depending on the clinical situation of ovarian cancer patients: upfront, platinum-sensitive and platinum-resistant relapse.


Le cancer de l'ovaire est la première cause de décès par cancer gynécologique. La plupart des patientes sont diagnostiquées au stade de carcinose péritonéale qui représente un défi thérapeutique. Sa prise en charge chirurgicale implique une cytoréduction maximaliste. Au cours des 30 dernières années, plusieurs stratégies de chimiothérapie intrapéritonéale ont été testées afin d'améliorer le contrôle de la carcinose péritonéale. Il s'agit des chimiothérapies intrapéritonéale adjuvante utilisée surtout en Amérique du Nord, hyperthermique intrapéritonéale (CHIP) et intrapéritonéale pressurisée en aérosols (PIPAC). Dans cet article, nous reprenons les données de la littérature sur chacune de ces trois approches thérapeutiques et proposons des algorithmes décisionnels selon la situation clinique des patientes traitées pour un cancer de l'ovaire : au diagnostic, récidive platine-sensible et platine-résistante.


Subject(s)
Carcinoma , Hyperthermia, Induced , Ovarian Neoplasms , Peritoneal Neoplasms , Algorithms , Antineoplastic Combined Chemotherapy Protocols , Carcinoma/therapy , Cytoreduction Surgical Procedures , Female , Humans , Hyperthermic Intraperitoneal Chemotherapy , Neoplasm Recurrence, Local/therapy , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/surgery
5.
J Clin Oncol ; 39(30): 3364-3376, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34339289

ABSTRACT

PURPOSE: In 2016, Kaiser Permanente Northern California regionalized gastric cancer care, introducing a regional comprehensive multidisciplinary care team, standardizing staging and chemotherapy, and implementing laparoscopic gastrectomy and D2 lymphadenectomy for patients eligible for curative-intent surgery. This study evaluated the effect of regionalization on outcomes. METHODS: The retrospective cohort study included gastric cancer cases diagnosed from January 2010 to May 2018. Information was obtained from the electronic medical record, cancer registry, state vital statistics, and chart review. Overall survival was compared in patients with all stages of disease, stage I-III disease, and curative-intent gastrectomy patients using annual inception cohorts. For the latter, the surgical approach and surgical outcomes were also compared. RESULTS: Among 1,429 eligible patients with gastric cancer with all stages of disease, one third were treated after regionalization, 650 had stage I-III disease, and 394 underwent curative-intent surgery. Among surgical patients, neoadjuvant chemotherapy utilization increased from 35% to 66% (P < .0001), laparoscopic gastrectomy increased from 18% to 92% (P < .0001), and D2 lymphadenectomy increased from 2% to 80% (P < .0001). Dissection of ≥ 15 lymph nodes increased from 61% to 95% (P < .0001). Surgical complication rates did not appear to increase after regionalization. Length of hospitalization decreased from 7 to 3 days (P < .001). Overall survival at 2 years was as follows: all stages, 32.8% pre and 37.3% post (P = .20); stage I-III cases with or without surgery, 55.6% and 61.1%, respectively (P = .25); and among surgery patients, 72.7% and 85.5%, respectively (P < .03). CONCLUSION: Regionalization of gastric cancer care within an integrated system allowed comprehensive multidisciplinary care, conversion to laparoscopic gastrectomy and D2 lymphadenectomy, increased overall survival among surgery patients, and no increase in surgical complications.


Subject(s)
Cancer Care Facilities/organization & administration , Carcinoma/therapy , Delivery of Health Care, Integrated/organization & administration , Gastrectomy/statistics & numerical data , Stomach Neoplasms/therapy , Adolescent , Adult , Aged , Aged, 80 and over , California , Carcinoma/secondary , Delivery of Health Care, Integrated/standards , Female , Gastrectomy/adverse effects , Gastrectomy/methods , Humans , Laparoscopy/statistics & numerical data , Length of Stay/statistics & numerical data , Lymph Node Excision/statistics & numerical data , Male , Middle Aged , Neoadjuvant Therapy/statistics & numerical data , Retrospective Studies , Stomach Neoplasms/pathology , Survival Rate , Treatment Outcome , Young Adult
6.
BMJ Case Rep ; 14(1)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33431443

ABSTRACT

Pubic osteomyelitis is a rare and often late-onset complication of radiation therapy and surgery for vulvar and vaginal carcinoma. It typically presents with vulvar pain, fever, vaginal discharge and/or gait disorders. Pubic osteomyelitis is often accompanied by fistulas or wound dehiscence in the pelvic area. Its accurate diagnosis and treatment are challenging and require a multidisciplinary team effort. In our patients, multiple combined surgical procedures, long-term antibiotic treatment and days to weeks of hospital admission were necessary to treat pubic osteomyelitis. We emphasise the importance of timely and adequate diagnosis and multidisciplinary approach resulting in a course of treatment that is as effective as possible, limiting the impact on quality of life, which is generally high in this group of patients.


Subject(s)
Carcinoma/therapy , Chemoradiotherapy, Adjuvant/adverse effects , Osteomyelitis/therapy , Radiation Injuries/therapy , Surgical Wound/therapy , Vulvar Neoplasms/therapy , Adult , Anti-Bacterial Agents/therapeutic use , Arthrodesis , Bone Transplantation , Carcinoma/pathology , Female , Humans , Leeching , Magnetic Resonance Imaging , Middle Aged , Osteomyelitis/diagnosis , Osteomyelitis/etiology , Patient Care Team , Pubic Bone/diagnostic imaging , Pubic Bone/radiation effects , Pubic Bone/surgery , Radiation Injuries/diagnosis , Radiation Injuries/etiology , Sacroiliac Joint/diagnostic imaging , Sacroiliac Joint/radiation effects , Sacroiliac Joint/surgery , Skin Transplantation , Surgical Wound/complications , Treatment Outcome , Vulva/pathology , Vulva/surgery , Vulvar Neoplasms/pathology
7.
Oncology (Williston Park) ; 34(6): 196-201, 2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32609864

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a disease marked by high rates of mortality; it is mostly incurable at the time of diagnosis. Only about 7% of patients survive 5 years after diagnosis. Diagnosis at a late stage and rapid progression with minimal response to available treatments are the main reasons for this poor outcome. It is crucial to identify individuals at high risk of developing PDAC so preventive and early detection measures can be employed. Approximately 10% to 15% of PDAC cases have a hereditary or familial basis. In the majority of PDAC cases, no main causative gene has been identified, but several known germline pathogenic mutations have been shown to be related to an increased risk of this cancer. The presence of 2 or more patients with pancreatic cancer within the circle of first-degree relatives, without the presence of a causative germline mutation, is defined as familial pancreatic cancer; this accounts for 4% to 10% of PDAC. Based on the growing evidence supporting the benefit of germline genetic testing in patients with PDAC, both the American Society of Clinical Oncology and the National Comprehensive Cancer Network recently updated their guidelines to include recommendations around genetic testing for patients with pancreatic cancer. However, there is no general consensus on the group of patients and individuals who should be studied and screened. We present a demonstrative case and review the available data on hereditary and familial PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma/genetics , Germ-Line Mutation , Pancreatic Neoplasms/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/diagnostic imaging , Carcinoma/therapy , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Pancreatic Ductal/therapy , Genetic Testing/methods , Humans , Male , Neoplastic Syndromes, Hereditary/diagnostic imaging , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Pancreatectomy/methods , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/therapy , Risk Factors
10.
Can J Surg ; 63(1): E71-E79, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32080999

ABSTRACT

Background: Peritoneal recurrences after cytoreductive surgery (CRS) and heated intraperitoneal chemotherapy (HIPEC) for appendiceal and colorectal cancers are frequent. This study aimed to evaluate the safety, technical feasibility and perioperative and long-term outcomes of repeat CRS/HIPEC in patients with recurrent peritoneal carcinomatosis of colorectal and appendiceal origin. Methods: Data were collected from patients treated from 2000 to 2016 for recurrent peritoneal carcinomatosis from appendiceal or colorectal cancer with CRS/HIPEC at 2 specialist centres. Data on demographics, procedure details, morbidity and survival were recorded. Analyses compared the iterations of CRS/HIPEC to assess the safety and effectiveness of repeat surgery. Results: Of all patients who underwent CRS/HIPEC in the 2 centres, 37 patients underwent a repeat procedure. Operative time was similar for the first and second surgeries (412.1 v. 412.5 min, p = 0.74) but patients had a significantly lower peritoneal carcinoma index score with the second surgery (21.8 in the first iteration v. 9.53 in the second iteration, p < 0.001) and significantly less blood loss (1762 mL in the first iteration v. 790 mL in the second iteration, p = 0.001). There was a nonsignificant decrease in grade III­IV complications and there was no 30-day mortality associated with repeat procedures. For patients with colorectal cancer, median disease-free survival was 9.6 months and median overall survival was 40 months. For patients with appendiceal cancer, median disease-free survival was 15 months and overall survival was 64.4 months. Conclusion: Repeat CRS/HIPEC procedures for recurrent appendiceal and colorectal peritoneal carcinomatosis are safe in well-selected patients, without increased morbidity or mortality, and they are associated with significant long-term survival, particularly for patients with appendiceal cancers. These results support the use of repeat CRS/HIPEC in these patients.


Contexte: Les récurrences péritonéales après une chirurgie cytoréductrice (CCR) et une chimiothérapie hyperthermique intrapéritonéale (CHIP) pour les cancers de l'appendice et colorectaux sont fréquentes. Cette étude visait à évaluer l'innocuité, la faisabilité technique et les résultats périopératoires et à long terme d'une reprise de CCR/CHIP chez les patients qui présentent une récurrence de carcinomatose péritonéale ayant son origine au niveau colorectal ou de l'appendice. Méthodes: Des données ont été recueillies sur des patients traités entre 2000 et 2016 pour une récurrence de carcinomatose péritonéale ayant son origine au niveau colorectal ou de l'appendice par CCR/CHIP dans 2 centres spécialisés. On a tenu compte des données démographiques, des détails des interventions, ainsi que de la morbidité et de la survie. Des analyses ont permis de comparer les premières et deuxièmes CCR/CHIP pour évaluer l'innocuité et l'efficacité des chirurgies répétées. Résultats: De tous les patients soumis à des CCR/CHIP dans les 2 centres, 37 ont subi l'intervention de nouveau. Le temps opératoire a été similaire pour les premières et les deuxièmes chirurgies (412,1 c. 412,5 min, p = 0,74), mais les patients présentaient un score de carcinomatose péritonéale beaucoup plus bas lors de la deuxième chirurgie (21,8 pour la première intervention c. 9,53 pour la seconde, p < 0,001) et des pertes sanguines significativement moindres (1762 mL pour la première intervention c. 790 mL pour la seconde, p = 0,001). On a noté une diminution non significative des complications de grades III­IV et on n'a déploré aucune mortalité à 30 jours en lien avec la reprise de l'intervention. Pour les patients atteints d'un cancer colorectal, la survie médiane sans maladie a été de 9,6 mois et la survie médiane globale a été de 40 mois. Pour les patients atteints d'un cancer de l'appendice, la survie médiane sans maladie a été de 15 mois et la survie médiane globale a été de 64,4 mois. Conclusion: La reprise des CCR/CHIP pour les récurrences de carcinomatose péritonéale ayant leur origine au niveau colorectal ou de l'appendice est sécuritaire chez les patients soigneusement sélectionnés, sans accroissement de la morbidité ou de la mortalité, et elles sont associées à une survie à long terme significative, particulièrement chez les patients ayant un cancer de l'appendice. Ces résultats appuient la reprise des CCR/CHIP chez ces patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Appendiceal Neoplasms/therapy , Carcinoma/therapy , Colorectal Neoplasms/therapy , Cytoreduction Surgical Procedures , Hyperthermia, Induced , Neoplasm Recurrence, Local/therapy , Outcome Assessment, Health Care , Peritoneal Neoplasms/therapy , Reoperation , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Appendiceal Neoplasms/mortality , Appendiceal Neoplasms/pathology , Canada/epidemiology , Carcinoma/mortality , Carcinoma/secondary , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Combined Modality Therapy , Cross-Sectional Studies , Cytoreduction Surgical Procedures/adverse effects , Cytoreduction Surgical Procedures/mortality , Feasibility Studies , Female , Humans , Hyperthermia, Induced/adverse effects , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Reoperation/adverse effects , Reoperation/mortality , Retrospective Studies
11.
Theranostics ; 10(1): 166-178, 2020.
Article in English | MEDLINE | ID: mdl-31903113

ABSTRACT

Organic chromophores have been well developed for multimodality imaging-guided photothermal therapy (PTT) due to their outstanding optical properties and excellent designability. However, the theranostic efficiencies of most currently available organic chromophores are restricted intrinsically, owing to their poor photostability or complex synthesis procedures. These drawbacks not only increase their cost of synthesis, but also cause side effects in PTT. Method: We presented a facile strategy for constructing a near-infrared (NIR)-absorbing perylenediimide structured with pH-responsive piperazine ring at the bay region. The chromophore was conjugated with carboxyl-end-capped PEG as side chains that can self-assemble into nanoparticles (NPs) in aqueous solution. The NIR optical properties and photothermal conversation ability of PPDI-NPs were investigated. We then studied the imaging-guided PTT of PPDI-NPs under NIR light illumination in 4T1 cells and mice respectively. Results: The excellent photostable PPDI-NPs had near-infrared fluorescence (NIRF) emission and high photothermal conversion efficiency in acidic microenvironment. Importantly, PPDI-NPs can be utilized for the precise detection of tumors by NIRF/photoacoustic/thermal trimodality imaging. Efficient PTT of PPDI-NPs was applied in vitro and in vivo with high biosafety. Conclusion: In summary, we developed pH-responsive perylenediimide nanoparticles as multifunctional phototheranostic agent with high stability and simple synthesis procedures. This study offers a promising organic chromophore for developing phototheranostics in cancer therapy.


Subject(s)
Breast Neoplasms , Carcinoma , Imides/therapeutic use , Perylene/analogs & derivatives , Photothermal Therapy , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/therapy , Carcinoma/diagnostic imaging , Carcinoma/therapy , Cell Line, Tumor , Female , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Multimodal Imaging , Nanoparticles , Perylene/therapeutic use , Photoacoustic Techniques , Theranostic Nanomedicine
12.
ACS Biomater Sci Eng ; 6(9): 5281-5289, 2020 09 14.
Article in English | MEDLINE | ID: mdl-33455277

ABSTRACT

Peritoneal seeding represents one of the most frequent sites of metastasis for late-stage gastrointestinal and gynecological cancer. At present, the major treatment method for peritoneal metastatic carcinoma (PMC) is the combination of cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC). Nevertheless, the 5 year survival rate of patients after these treatments is still far from satisfactory. Here, we report a biodegradable implant co-loaded with doxorubicin (DOX) and anti-PD-1 monoclonal antibody (aPD-1) (BI@DOX+aPD-1) for a combination of immunogenic chemotherapy and immune checkpoint therapy for PMC postoperative treatment. The bio-implant is fabricated with oxidized dextran (ODEX) and 4-arm poly(ethylene glycol) amine (4-arm PEG-NH2) by Schiff's base reaction at mild conditions, with DOX and aPD-1 loaded inside during and after the fabrication process, respectively. In vitro studies confirmed the slow and sustained release of DOX and aPD-1 from the bio-implants. In vivo studies showed that the bio-implants could be gradually degraded and maintain relatively high concentrations of therapeutic agents in the mouse abdomen. In a murine CT26 PMC model, the BI@DOX+aPD-1 resulted in a 89.7% tumor-suppression rate after peritoneal implantation. Importantly, the combination therapy of DOX and aPD-1 in the bio-implant showed an excellent synergistic effect with a Q value of 2.35. This easy-fabricated bio-implant combined with DOX and aPD-1 should be promising for clinical PMC postoperative treatment.


Subject(s)
Carcinoma , Hyperthermia, Induced , Peritoneal Neoplasms , Absorbable Implants , Animals , Carcinoma/therapy , Cytoreduction Surgical Procedures , Humans , Mice , Peritoneal Neoplasms/drug therapy
13.
Surg Today ; 50(2): 171-177, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31363844

ABSTRACT

PURPOSE: Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS + HIPEC) is the active treatment for peritoneal carcinomatosis of appendiceal origin. However, surgical management is sometimes difficult in patients with a high-tumor burden. METHODS: A high-tumor burden was defined as a peritoneal cancer index (PCI) ≥ 28. Among 49 patients receiving CRS + HIPEC, 29 had a PCI ≥ 28. RESULTS: Complete cytoreduction (CC-0/1) was achieved in 20 of the 29 patients with a PCI ≥ 28 and in all 20 patients with a PCI < 28. Among the patients achieving CC-0/1, gastrectomy or total colectomy was performed more frequently, the hospital stay was longer and postoperative complications were more frequent in those with a PCI ≥ 28 than in those with a PCI < 28. If CC-0/1 was achieved, the overall survival was comparable between patients with a PCI ≥ 28 and a PCI < 28. However, the recurrence-free survival was significantly worse for patients with a PCI ≥ 28 than for those with a PCI < 28 (5-year survival: 73.7% vs. 5.9%). Patients with recurrence who underwent repeat CRS showed a better overall survival than those without repeat CRS. Among patients with a PCI ≥ 28, a performance status (PS) of 2/3 was a significant prognostic factor (hazard ratio = 5.132). CONCLUSIONS: In patients with a high-tumor burden undergoing CRS + HIPEC, postoperative complications were more frequent, and the recurrence rate was higher than in those without a high-tumor burden. Repeat CRS improved the survival of patients with recurrence. The PS was a key indicator when selecting patients suitable for aggressive resection.


Subject(s)
Appendix , Carcinoma/surgery , Peritoneal Neoplasms/surgery , Antineoplastic Agents/administration & dosage , Carcinoma/mortality , Carcinoma/pathology , Carcinoma/therapy , Cytoreduction Surgical Procedures/methods , Humans , Hyperthermia, Induced , Neoplasm Recurrence, Local , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/therapy , Survival Rate
14.
Eur J Surg Oncol ; 46(4 Pt A): 577-581, 2020 04.
Article in English | MEDLINE | ID: mdl-31677939

ABSTRACT

INTRODUCTION: Morbidity associated with cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) is due to the synergistic effect of cytoreduction, effect hyperthermia and the cytotoxic agents used for HIPEC. This study was done to analyse the postoperative morbidity in relation to the chemotherapy agent used in patients undergoing CRS-HIPEC for peritoneal surface malignancy (PSM) in Indian set up. MATERIALS AND METHODS: Patient with PSM, underwent CRS-HIPEC as per the institutional protocol. Patients were stratified as per the chemotherapy drug used during HIPEC & perioperative outcome were documented. RESULTS: 163 patients underwent CRS-HIPEC for PSM: 67.4% were of ovarian primary. Others were colorectal, appendicular, gastric primary and rare tumors.Cisplatin was the most common drug used: as alone (57.05%) or in combination with Adriamycin (12.88%). Mitomycin-C (MMC) was used in 20% and oxaliplatin in 10%.Grade 3-5 morbidity in the whole cohort was 44.8% and grade 1-2 was 74%.Grade 1-2 electrolyte abnormality was the most common morbidity overall and grade 3-4 hematological toxicity was the most common severe morbidity. Frequency of grade 3-5 morbidity were 38.7%, 48.5%,50% and 61.9% for Cisplatin alone, MMC, oxaliplatin and Adriamycin + cisplatin respectively. None of the patients had grade 3-4 nephrotoxicity as sole complication. All major complications were highest in the group who received Adriamycin. Cisplatin was associated with higher rate of electrolyte imbalance, oxaliplatin with post-operative bleeding. Rates of other complications did not differ significantly. CONCLUSION: Cisplatin followed by MMC were the well tolerated drugs during HIPEC and tolerance to Adriamycin combination regimen in Indian patients was poor.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma/therapy , Cytoreduction Surgical Procedures , Hyperthermia, Induced , Peritoneal Neoplasms/therapy , Postoperative Complications/epidemiology , Adult , Aged , Appendiceal Neoplasms/pathology , Carcinoma/secondary , Cisplatin/administration & dosage , Cisplatin/adverse effects , Colorectal Neoplasms/pathology , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Humans , India/epidemiology , Male , Middle Aged , Mitomycin/adverse effects , Ovarian Neoplasms/pathology , Oxaliplatin/adverse effects , Peritoneal Neoplasms/secondary , Postoperative Complications/chemically induced , Postoperative Hemorrhage/chemically induced , Postoperative Hemorrhage/epidemiology , Stomach Neoplasms/pathology , Water-Electrolyte Imbalance/chemically induced , Water-Electrolyte Imbalance/epidemiology
15.
Eur J Surg Oncol ; 46(4 Pt A): 675-683, 2020 04.
Article in English | MEDLINE | ID: mdl-31806517

ABSTRACT

The presence of peritoneal metastases in patients with advanced colorectal cancer is associated with poor prognosis but the mechanisms for this are unclear. This review summarises the current knowledge of the pathophysiology, clinical features, prevalence, prognosis, and molecular biology of peritoneal metastases and the risk factors for the development of peritoneal metastases following resection of a primary colorectal tumour. Furthermore, the evidence for treatment strategies are described including cytoreductive surgery, hyperthermic intraperitoneal chemotherapy, early post-operative intraperitoneal chemotherapy, sequential post-operative intraperitoneal chemotherapy and emerging novel strategies. Active areas of research should include the identification of individuals at high risk of peritoneal metastases after curative resection of primary tumour, development of a surveillance program for high-risk patients, optimisation of systematic therapies and further investigation of the use of intraperitoneal chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/therapy , Colorectal Neoplasms/pathology , Cytoreduction Surgical Procedures , Peritoneal Neoplasms/therapy , Carcinoma/genetics , Carcinoma/secondary , Chemotherapy, Adjuvant , Colorectal Neoplasms/genetics , Humans , Hyperthermia, Induced , Immunotherapy, Adoptive , Infusions, Parenteral , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Risk Factors
16.
Eur J Surg Oncol ; 46(4 Pt A): 590-599, 2020 04.
Article in English | MEDLINE | ID: mdl-31806520

ABSTRACT

BACKGROUND: The peritoneal cancer index (PCI) calculated during exploratory laparotomy is a strong prognostic factor for overall survival (OS) in patients with colorectal peritoneal metastases (PM) who undergo cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS + HIPEC). Progression of the PCI between diagnostic laparoscopy (DLS) and potential CRS + HIPEC (ΔPCI) might be a more dynamic prognostic factor for OS after CRS + HIPEC. MATERIALS AND METHODS: Between 2012 and 2018, all colorectal PM patients who underwent an exploratory laparotomy for potential CRS + HIPEC after DLS were retrospectively identified from a prospectively maintained database. Patients were divided into stable disease (ΔPCI 0-3), mild progression (ΔPCI 4-9), or severe progression (ΔPCI ≥10). Kaplan-Meier analysis and a multivariate Cox regression were performed. RESULTS: Eighty-four patients (ΔPCI 0-3, n = 35; ΔPCI 4-9, n = 34; and ΔPCI ≥10, n = 15) were analysed. Median OS after CRS + HIPEC was significantly decreased in patients with a ΔPCI of 4-9 (35.1 [95% CI 25.5-44.6]) or ΔPCI ≥10 (24.1 [95% CI 11.7-36.5]) compared to patients with a ΔPCI of 0-3 (47.9 [95% CI 40.0-55.7], p = 0.004). In multivariate regression analysis, ΔPCI remained an independent risk factor for OS: ΔPCI 4-9 HR 3.1 (95% CI 1.4-7.2, p = 0.007) and ΔPCI ≥10 HR 4.4 (95% CI 1.5-13.1, p = 0.007). CONCLUSION: A high ΔPCI is an independent dynamic prognostic factor for OS and might reflect a more aggressive tumour biology in patients with colorectal PM. HIPEC surgeons should be aware of a high-ΔPCI-associated diminished prognosis and should reconsider CRS + HIPEC when confronted with a ΔPCI ≥10.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma/secondary , Colorectal Neoplasms/pathology , Mitomycin/therapeutic use , Peritoneal Neoplasms/secondary , Aged , Carcinoma/therapy , Cytoreduction Surgical Procedures , Disease Progression , Female , Humans , Hyperthermia, Induced , Kaplan-Meier Estimate , Laparotomy , Male , Middle Aged , Peritoneal Neoplasms/therapy , Prognosis , Proportional Hazards Models , Survival Rate , Tumor Burden
17.
Med Arch ; 73(5): 331-337, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31819307

ABSTRACT

INTRODUCTION: Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy (CRS ± HIPEC) has positive effects on the survival of patients with peritoneal carcinomatosis (PC) due to intra-abdominal tumors. Currently, the available literature on the safety of the Enhanced Recovery After Surgery (ERAS) protocol for PC, which is associated with severe morbidity and mortality, is insufficient. AIM: This study aimed to present our results from treating patients using the ERAS protocol for PC that developed due to intra-abdominal tumors. MATERIAL AND METHODS: The data of 120 consecutive patients with PC due to different etiologies of abdominal origin and who underwent CRS ± HIPEC were analyzed. The patients were divided into two groups according to whether the ERAS protocol was applied. Information on demographics, length of hospital stay, cost, morbidity, and mortality was statistically compared between groups. RESULTS: A total of 102 patients were included in the study. The first 40 patients did not undergo the ERAS protocol, whereas 62 patients did undergo the protocol. The mean length of hospital stay was 10 days in the non-ERAS group and 7 days in the ERAS group. The ERAS group was observed to have earlier mobilization, earlier gas and stool release, lower oral intake, and fewer respiratory problems than the non-ERAS group. CONCLUSION: CRS ± HIPEC has a positive effect on survival. The simultaneous application of the ERAS protocol with the aforementioned procedure has positive effects on intestinal motility and postoperative outcomes. In addition, this protocol may reduce costs by shortening the length of hospital stay.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma/therapy , Cytoreduction Surgical Procedures/methods , Enhanced Recovery After Surgery , Hyperthermia, Induced/methods , Peritoneal Neoplasms/therapy , Sarcoma/therapy , Adult , Aged , Carcinoma/secondary , Colorectal Neoplasms/pathology , Early Ambulation , Female , Gastrointestinal Motility , Humans , Infusions, Parenteral , Length of Stay , Male , Middle Aged , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Postoperative Complications/epidemiology , Sarcoma/secondary , Stomach Neoplasms/pathology
19.
J Visc Surg ; 156(5): 377-379, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31466831

ABSTRACT

When peritoneal metastases are diagnosed (strong agreement of experts): (i) seek advice from a multidisciplinary coordination meeting (MCM) with large experience in peritoneal disease (e.g. BIG RENAPE network); (ii) transfer (or not) the patient to a referral center with experience in hyperthermic intraperitoneal chemotherapy (HIPEC), according to the advice of the MCM. With regard to systemic chemotherapy (strong agreement of experts): (i) it should be performed both before and after surgery, (ii) for no longer than 6 months; (iii) without postoperative anti-angiogenetic drugs. With regard to cytoreductive surgery (strong agreement of experts): (i) Radical surgery requires a xiphopubic midline incision; (ii) no cytoreductive surgery via laparoscopy. With regard to HIPEC: HIPEC can be proposed for trials outside an HIPEC referral center (weak agreement between experts): (i) if surgery is radical; (ii) if the expected morbidity is "reasonable"; (iii) if the indication for HIPEC was suggested by a MCM, and; (iv) mitomycin is preferred to oxaliplatin (which cannot be recommended) for this indication.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/secondary , Chemotherapy, Cancer, Regional Perfusion/methods , Colorectal Neoplasms/pathology , Cytoreduction Surgical Procedures/methods , Hyperthermia, Induced/methods , Peritoneal Neoplasms/secondary , Antineoplastic Agents/therapeutic use , Carcinoma/therapy , Chemotherapy, Cancer, Regional Perfusion/standards , Combined Modality Therapy , Cytoreduction Surgical Procedures/standards , Humans , Hyperthermia, Induced/standards , Peritoneal Neoplasms/therapy
20.
Theranostics ; 9(10): 2897-2909, 2019.
Article in English | MEDLINE | ID: mdl-31244931

ABSTRACT

Inaccessibility of deep-seated malignant cells in the central region of tumors and uncontrollable tumor recurrence represent a significant challenge for conventional synergistic cancer therapy. Herein, we designed a novel nanoplatform based on hierarchical drug release for deep cascade cancer therapy including localized photothermal therapy, systematic chemotherapy, and elicited immune responses. Methods: The first-step chemotherapy could be carried out by polydopamine (PDA) releasing doxorubicin (DOX) in the specific microenvironment of lysosomes (pH 5.5). The branched gold nanoshells and PDA converted the light to heat efficiently to accomplish the second-step photothermal therapy and collapsed biomimetic vesicles (BVs) to release paclitaxel (PTX), which promoted the third-step of chemotherapy and triggered immune responses. Results: After 10 days of treatment, there were no obvious residual tumors in tumor-bearing mice. Significantly, 10 days after stopping treatment, mice in the drug immune-therapeutic group showed little tumor recurrence (1.5 times) compared to substantial recurrence (20 times) in the conventional treatment group. Conclusion: The hierarchical drug release and cascade therapeutic modality enhance the penetration of drugs deep into the tumor tissue and effectively inhibit recurrence. This cascade therapeutic modality provides a novel approach for more effective cancer therapy.


Subject(s)
Carcinoma/therapy , Drug Delivery Systems , Drug Therapy/methods , Hyperthermia, Induced/methods , Immunotherapy/methods , Nanocomposites/administration & dosage , Phototherapy/methods , Animals , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Doxorubicin/administration & dosage , Drug Liberation , Gold/administration & dosage , HeLa Cells , Humans , Immunologic Factors/administration & dosage , Mice , Models, Biological , Neoplasm Transplantation , Paclitaxel/administration & dosage , Photosensitizing Agents/administration & dosage , Transplantation, Heterologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL