Abstract
Intraperitoneal chemotherapy has demonstrated significant pharmacologic and clinical advantage over traditional intravenous administration for cancers that are restricted to the peritoneal cavity. The combination of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC) has become the standard technique used to fight ovarian and gastrointestinal cancers in many centers. However, challenges remain for HIPEC to contact the entire peritoneal surface, penetrate the tumor tissue, and transport to the lymphatics and other metastatic sites. New innovations in delivery technique, such as heated aerosol, and in delivery molecules, such as microparticles, nanoparticles, nanogels, and tumor-penetrating peptides are being tested in animal models and will likely soon be in human trials. Improvements in overall care, such as the recent clinical trial of an oral agent for maintenance therapy in ovarian carcinoma, will continue in this field for the next 20 years.
Introduction
Despite advances over the past 20 years in treatment options for metastatic ovarian or gastrointestinal cancers, survival is relatively poor. Ovarian cancer is the sixth most common neoplasm in women [1], and local recurrence of stomach, pancreas, colo-rectal cancers varies between 30 % and 60 % [2–6]. The chief strategy that is employed at present is a combination of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) [1, 6]. HIPEC is a regional treatment involving a heated solution in the peritoneal cavity and high drug concentrations [6, 7]. HIPEC is often performed immediately after peritonectomy and visceral resection to remove cancer that can be detected. Variations of this treatment such as use of a heated aerosol [8] are used before cytoreductive surgery and HIPEC. Tumor cells from the primary cancer can invade the serosa or local blood vessels or migrate to the local lymphatics that subsequently drain from the intestinal tract to cisterna chyli or directly from the peritoneal cavity to the sub-diaphragmatic lymphatics. Treatment solutions in the peritoneal cavity can penetrate not only the local tissue but also these same lymphatic routes. The presence of ascites may indicate that these lymphatics are blocked by the tumor cells. To the extent that HIPEC: (a) covers the peritoneal surface, (b) penetrates the tissue, and (c) transports the active agent to the lymphatics, the technique treats the residual cancer and prevents peritoneal metastases of the primary malignancy. These factors are the technical challenge and depend on the molecular size of the agent used, the peritoneal area in contact with the solution (volume and mixing conditions of the solution), the intraperitoneal pressure of the solution (important for penetration of larger molecular sizes), and the microenvironment of the tumor (i. e., interstitial pressure). This paper will review the state of the art and technical innovations in delivery of agents and in the agents themselves.
Theory
As discussed in our previous publication [9], the simplest model to estimate the mass transfer of a small (MW<6,000 Da), non-metabolized, water soluble drug across the peritoneum is the following:
where PA=the mass transfer-area coefficient (see [10, 11]) and CP=concentration in the peritoneal cavity, CB=concentration in the blood.
We extended that equation to the description of the free drug in the normal extracellular space (Ce) to be:
where D=diffusivity of the drug in the extracellular space and k=rate constant of removal of the drug by the circulation (see reference [9]). The term “normal” typically means that blood vessels are distributed approximately evenly throughout the tissue space and extracellular tissue pressure in the range of –1 to 0 mmHg. This simple model provided estimates of the penetration (to a low point of 5 % of the peritoneal concentration) of an EDTA-sized molecule at approximately 400–500 μm in the gastrointestinal tract of the rat [12]. This is perhaps the most significant reason for cytoreductive surgery: theoretically, penetration of a small agent can be less than 1 mm.
However, there are a variety of factors that will alter this estimate [13]. An irregularly distributed vasculature of a tumor can affect the properties of the interstitium or microenvironment and can alter the diffusive transport of a small molecule. The drug will likely be taken up by tumor cells and metabolized, and the concentration could drop off rapidly and be difficult to estimate [14]. The total drug taken up by a tumor for could be significantly enhanced over time: Los et al. [15, 16] measured the platinum distribution in CC531 colon adenocarcinoma implanted in the rat peritoneum and determined that concentrations were significantly elevated at 1 mm from the peritoneum after 24–48 h. Others have shown that the penetration depth for most alkylating agents is on the order of 1–3 mm [17].
Au et al. [18] has developed a sophisticated multiscale tumor, spatiokinetic model for intraperitoneal therapy, which assumes contact of the treating solution with the tumor surface. This effort models paclitaxel, both free and drug bound to soluble proteins, the transfer from the cavity via lymphatic drainage, transperitoneal transport, and subperitoneal tumors and tissue, as well as the disposition in the systemic compartment with final elimination. The tumor is modeled with a variable cell density, and transcellular and transvascular transport are included within the interstitium. Model output is fitted to in vivo data of 6 h 3H-paclitaxel autoradiography with densitometric standards. A subsequent sensitivity analysis demonstrated that parameters such as tumor interstitial diffusivity, vessel surface area per unit tissue volume, and maximum cellular binding capacity were the most important to model output. This model predicts penetration of the agent given that the agent is in contact with the tumor for a specified amount of time. Others have made efforts to model or measure the penetration of macromolecules into tumors [19–22].
Area of contact
Of paramount importance to intraperitoneal therapy is the area of contact of the chemotherapeutic solution with all potential targets. Sugarbaker [23] infused blue dye into the peritoneal cavity and demonstrated that intraperitoneal fluid distribution and contact with the peritoneum was quite variable, likely due to post-operative adhesions and folds of the peritoneum in contact with each other. To correct for this, a technique of expansion is used to reach as many surfaces; Sugarbaker [24] introduced the “coliseum technique” that involved attaching the skin of the abdomen to a retractor ring that was suspended above the laparotomy wound. A plastic sheet covered the wound to allow the surgeon’s hand to stir the solution in the cavity in order to achieve a more uniform exposure of the solution throughout the cavity. Others developed a “peritoneal access device” to achieve maximum exposure of the solution to the peritoneal surface [25, 26]. While both of these techniques help to distribute the fluid throughout the cavity, the loss of heat from the open wound resulted in decreased hyperthermia effects and creation of a toxic aerosol in the operating theater. There were also portions of the parietal peritoneum that were not fully exposed and this could possibly create a site for tumor reoccurrence [27]. To remedy this, a closed system is used with a single inflow catheter and one or more outflow catheters [28, 29]. While the closed system allows better control of the heat and perfusion, it may re-introduce tumor cells back into the cavity [29] and decrease access of the solution to all peritoneal areas, even limiting fluid contact with some stirring techniques.
In a recent paper, Lotti et al. [30] discuss laparoscopic HIPEC, which is proposed as a bridge between open and closed techniques. After cytoreductive surgery, four Jackson–Pratt drains are inserted into the abdominal cavity (right subphrenic, right inferior, below the left hemidiaphragm, left pelvis) to provide for outflow of the treatment fluid. A separate catheter is inserted into the left flank to measure the intra-abdominal pressure, along with two thermal probes. The abdominal wound is closed with multiple locking sutures with three balloon trocars at the junction of the sutures, with the upper trocar connected to the HIPEC inflow tube, the middle trocar to the heated CO2 insufflator to establish pneumoperitoneum (intraperitoneal pressure=12 mmHg), and the lower trocar is connected to the smoke evacuator device. The upper trocar is used for inflow and laparoscopic viewing of the peritoneum. The abdominal contents are stirred by two palpators via the middle and lower trocars, with the cycle being 90 min of perfusion alternating with 5 min of stirring and 10 min of perfusion. After the procedure, the trocars, pressure catheter, and thermal probe are removed, and the wound is reopened, as required. While complicated, the procedure is proposed to reach the entire peritoneum, including the abdominal wall.
Effect of heat
Intraperitoneal therapy can be carried out with or without heat. Spratt et al. carried out some of the earliest research into this technique in 1980 [31, 32]. This group first demonstrated the technique in dogs, and subsequently reported its use in a patient with pseudomyxoma peritonei, after extensive resection of the tumor including the ascending colon, the transverse colon, the omentum, the spleen and the distal pancreas. Two catheters were placed in the cavity: one in the upper left quadrant with the end laid into the subphrenic region, while the second was placed in the lower right quadrant with the end on the pouch of Douglas. Methotrexate and thiotepa were infused with concentrations over 30 min in the cavity being 1,000 times those observed in the serum [23].
Optimal temperatures are typically between 42 and 43 °C, where synergy exists between heat and drug toxicity [33]. Hyperthermia of >42.0 °C induces alterations in the microenvironment and tumor blood flow that impair oxygen supply and induce acidosis. These effects likely combine to decrease the nutritive blood flow to the tumor. Most alkylating agents, such as cyclophosphamide and ifosfamide, nitrosources such as carmustine and lomustine, bleomycin, nucleoside analog (gemcitabine), and the platinum compounds are enhanced in their cytotoxic properties at temperatures above 40.5 °C, while 5-fluorouracil demonstrates no increased cytotoxicity with increased temperature [34].
The range of effects of hyperthermia on the cell includes: destabilizing the cell membrane, changes in cell shape, impaired transmembrane transport, changes in membrane potential and modulation of transmembrane efflux pumps, induction of apoptosis, impairment of protein synthesis, protein denaturation, aggregation of proteins at the nuclear matrix, induction of heat-sensitive protein synthesis, inhibition of enzyme repair, altered DNA conformation, alteration of intracellular metabolism of other substrates, and alteration of gene expression and signal transduction [17, 33]. However, thermo-tolerance has been observed with different forms of drug resistance [35].
Typically, the heated solution is infused into the peritoneal cavity immediately after cytoreductive surgery and circulated or allowed to dwell 60–120 min. This is followed by early post-operative intraperitoneal chemotherapy (EPIC) is begun in the first post-operative day and the solution dwells for 23 h, drained for 1 hour and then re-administered; the duration of therapy is typically 1 week [17]. Complications due to the heated solutions are due to bowel perforations due to surgical trauma. [26, 29, 36–40]. Systemic toxicity, especially renal failure due to platinum drugs and heat, is a major consideration [37–40].
Penetration of intraperitoneally delivered agent
Tissue penetration is a major impediment to the success of intraperitoneal delivery. In normal tissue, the interstitium is –1 to 0 mmHg [41], and penetration of small molecules (MW<1,000 Da) conforms to the typical diffusion gradient [11, 42, 43], while penetration of larger molecules is dependent on intraperitoneal pressure (typically 4–10 mmHg) [44–46]. In animal models [47, 48], Flessner demonstrated that some tumors exhibit high interstitial pressures (10–20 mmHg) that set up a pressure-driven convection outward from the tumor to the periphery. This creates a convective barrier to large solutes that depend on pressure to propel the molecules to the interior of the tumor and effectively blocks penetration to these tumors with high pressures [47–50]. Human tumors as well have high interstitial pressures as noted in many studies [51–55]. Small solutes may also be retarded in their transport, depending on the tumor interstitial fluid movement from the interior to the periphery [56]. Gremonprez et al. [57] demonstrated that pretreatment of the tumors with gavage-treatments with vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR) inhibitors decreases interstitial pressure (from 20 to 5 mmHg), increases penetration of oxaliplatin and slows tumor growth in a mouse colorectal cancer model.
Innovations in intraperitoneal chemotherapy
PIPAC: pressurized intraperitoneal aerosol chemotherapy
Among recent innovations in intraperitoneal drug delivery is PIPAC, the use of a novel clinical approach to enhance penetration of the drug, enhancement of the area covered, and delivery of the dose. This technique uses a probe placed through the abdominal wall and connected to a micropump to deliver the pressurized aerosol. Reymond et al. [58] tested the feasibility of a pneumoperitoneum in a large animal model using a microvaporisator, in an effort to overcome the limitations of intraperitoneal chemotherapy including direct penetration of drugs into tumor tissue and the unequal distribution throughout the cavity. They combined gas and pressure in order to overcome these problems and noted improved coverage of the entire surface of the peritoneum over and above the typical “peritoneal washing”. The group subsequently tested the concept in a box with an excised tumor section: they determined that under a pressure of 12 mmHg, penetration of the tracer material was 1 mm [59, 60].
Subsequent studies demonstrated in a Phase 2 study in women with ovarian carcinoma that the 30 min application of cisplatin and doxorubicin resulted in an objective tumor response in 76 % of the 34 patients treated with three treatments over 28–42 days with only a few serious adverse events [61–63]. Liver and renal toxicities appear to be minimal [64], and quality of life was maintained in a cohort of 91 patients [65, 66]. The most recent use of PIPEC has been as an adjuvant therapy in preparation for surgical cyto-reduction and HIPEC [8].
Jung et al. [66, 67] has tested a hyperthermic version of PIPAC in swine, which he has termed H-PAC. Surgery included placement of three trocars: one in the superior peritoneum, one just beneath the umbilicus, and one in the lower pelvis. Any of the trocars could be used for the laparoscope, and the mid-abdomen was considered the most favorable site to administer the solution as an aerosol spray. Insulating the heated CO2 line was found to be necessary to prevent loss of heat. The temperatures of the solution delivered were 38.7–40.8 °C, and the authors considered the experiments a success, although they did not check the area covered or the depth of penetration in the experimental tissues.
A second innovation in the PIPAC technique has been offered: electrostatic precipitation as an adjunct to pressurized intraperitoneal aerosol chemotherapy, termed ePIPAC [68]. After applying PIPAC [69] by inflating the peritoneum to 12 mmHg and inserting two balloon safety trocars into the abdomen and infusing the pressurized aerosol for approximately 30 min, an electric current was added for 30 min via an electrode in the abdomen. The negatively charged particles were then deposited on the positively charged peritoneal tissue. Toluidine blue was in the solution used to determine the tissue area distribution and noncoding small DNA fragments labeled with Cy5 (DT01) were used to determine tissue penetration. Results in pigs demonstrated spread of the solution to many parts of the peritoneum and somewhat better penetration with ePIPAC than standard PIPAC (p<0.06); there was almost no DT01 left in the peritoneal cavity after ePIPAC vs. PIPAC (p<0.01).
However, in a simulated ex vivo delivery at a pressure of 12 mmHg, Khosrawipour et al. demonstrated that the position of the tissue directly under the spray nozzle had a major impact on the concentration delivered to the tissue [70]. Penetration was significantly affected by drug (doxorubicin) dose but not by pressure [71]. Irradiation of the tissue prior to treatment did not enhance the penetration [72]. In a postmortem swine model [73], doxorubicin was aerosolized into the peritoneal cavity at a pressure of 12 mmHg and 32 °C, and the distribution and depth of penetration were determined a nine different positions in the cavity. These experiments demonstrated a variable penetration in all tissues, with the highest concentrations in the tissue closest to the pump probe.
Innovative agents for intraperitoneal chemotherapy
Lu et al. [13] have outlined optimal characteristics for the development of drug-loaded tumor penetrating microparticles:
Have long retention in the peritoneal cavity;
Selectively adhere to tumors;
Have deep tumor penetration;
Provide instantaneous and sustained drug release to obtain optimal peritoneal delivery.
Absorption is a passive process of diffusion and convection (with the proviso that the delivery pressure is greater than the tumor interstitial pressure). Molecules that are less than 20 kDa can easily cross the peritoneum [74], but larger substances (molecular size=50–200 nm) transport chiefly via the lymphatics and may be taken up by the lymph nodes [75]. Therefore, free drugs or nanoparticles that <50 nm are capable of absorption into the surrounding tissue. Smaller particles (<5 μm) distribute widely in the peritoneal cavity [13], while larger particles may collect in the lower part of the abdominal cavity [76]. Selective adherence of the microparticle is desirable; activated carbon particles have shown promise with certain sarcomas [77]. Tumor priming, such as use of a drug such as paclitaxel or doxorubicin to expand the tumor interstitial space [78–80] is useful in enhancing drug transport and drug penetration.
Preclinical agents
The determination of platinum in tumor tissue has been recently improved by Carlier et al. [81]. This group used a synchrotron radiation X-ray fluorescence spectrometry and laser ablation-inductively coupled plasma-mass spectrometry to measure platinum distribution and penetration in ovarian cancer peritoneal xenografts after intraperitoneal administration of cisplatin at two different temperatures. Using principal component analysis, they correlated several molecules with the distribution of platinum. The results demonstrated higher concentrations of platinum after hyperthermic perfusion and increased penetration. However there were no differences in platinum distribution between cells and stroma.
Tsai et al. [82] exposed mice to intraperitoneal paclitaxel in 3 formulations: Cremophor formulation, paclitaxel-loaded gelatin nanoparticles, and polymeric microparticles to evaluate their advantage in targeting and treatment of tumors. They utilized whole-body, quantitative autoradiography to determine the relative distribution in tissues and penetration, and found slower drug clearance with the microparticles, 5–22 times greater advantage, and twice the survival advantage. They [83] subsequently investigated the effects of smaller (dia=5–6 μm) vs. larger (50–60 μm) microparticles in a murine model of pancreatic cancer and found that the smaller particles were more evenly distributed in the peritoneal cavity and demonstrated longer survival and higher dose efficiency. Au et al. [84] has recently reviewed these preclinical results as a preliminary step in the use of drug-carrying microparticles in an investigational new drug (IND)-enabled study.
Paclitaxel can also be combined with mesoporous silica nanoparticles to treat ovarian carcinoma [85]. Subsequently, Fu et al. extended the results and used in vivo imaging in a murine model at three different times and then sacrificed and cryo-sectioned the animals to study the penetration of the nanoparticles [86]. This group used tumor bearing mice to compare IV with intraperitoneal administration of paclitaxel. Marked increase in cellular accumulation of paclitaxel occurred with silica nanoparticles vs drug alone.
Cisplatin-incorporating hyaluronan (HA) nanogels have recently been fabricated by mixing the drug with conjugating chelating ligands (iminodiacetic acid or malonic acid to HA) to form a cross-linked, cisplatin-loaded hyaluronan nanogels [87]. The HA gels selectively inhibited the growth of gastric cancer cells. In vivo experiments in a murine model demonstrated the HA nanogels specifically localized to the peritoneal nodules after intraperitoneal administration. Penetration assays showed a significantly higher ability to penetrate tumors than unconjugated hyaluronan.
Tumor-penetrating peptides are a class of new tools in treating metastatic peritoneal carcinoma that transport actively through tumor tissue [88]. These substances require a primary receptor, proteases, and neurophilins. These peptides bind to a tumor-specific receptor (e. g., integrins) and are then converted to bind neurophilin-1 or neurophilin-2′ this exposes a c-terminal amino acid sequence for the neurophilin (NRP) binding [89]. One such tumor-penetrating peptide, a 9-amino-acid-based cyclic, arginine-glycine-aspartic acid (RGD) has the necessary characteristics to enhance penetration of co-applied molecules into peritoneal tumors, no matter the degree of vascularity and improves the therapeutic index. Sugahara et al. [90, 91] demonstrated these properties with a murine model of disseminated gastric cancer with imaging and immunostaining of the harvested tissues. The animals were also co-injected with dextran and/or doxorubicin and demonstrated marked differences in those animals with the iRGD molecule with increased penetration (3–10 mm vs 0.5 mm) over the non-tumor penetrating peptide. iRGD also facilitated entry of dextran into both avascular and vascularized peritoneal tumors with intraperitoneal-coinjection. Other non-penetrating peptides did not facilitate dextran entry. In preparation for human studies, they showed similar properties in excised human tumors [92]. This group has demonstrated facilitation of tumor penetration with iRGD conjugated to polymerase in the treatment of colon or gastric cancer induced in murine models [93, 94].
Clinical innovations
Recurrent ovarian cancer is a serious problem that continues to challenge clinical researchers. Fagotti et al. obtained encouraging results in two Phase 2 studies on the use of HIPEC following secondary cytoreduction in patients with recurrent platinum-sensitive ovarian cancer [95, 96]. They subsequently carried out a case-control study in patients who had platinum-sensitive ovarian carcinoma with 30 cases and 37 controls and 24 months of follow-up [97]. They found that survival was significantly better at 2 years (96.7 vs 75.7 %) and at 5 years (68.4 vs. 42.7 %) with HIPEC (with the administration of oxaliplatin 460 mg/m2 at 41.5 °C) than with chemotherapy or surgery plus chemotherapy. This group [98, 99] screened 231 women between 2004 and 2015 with platinum-sensitive ovarian cancer. Approximately 83 % of the women had stage 3 or 4 disease as well as serous (85 %) and high-grade (93 %) ovarian cancer. Nearly all underwent primary debulking. The screening process reduced the population to 70 for recurrent disease with all undergoing secondary cytoreduction plus HIPEC with either cisplatin (38.6 %) or oxaliplatin (61.4 %); follow-up time ranged between 48 and 128 months with 34 deaths and 36 who survived. These findings demonstrate the effectiveness of HIPEC in platinum-sensitive ovarian cancer.
As detailed above, there is significant research in the use of nanoparticles as means of drug delivery to peritoneal malignancies. In a recent Phase 1 study, Williamson et al. [100] detailed findings of an open-label, dose-escalating examination of the safety, tolerability, pharmacokinetics and preliminary tumor response of Nanotax, a nanoparticulate formulation of paclitaxel. The particles had a diameter of 600–700 nm, with 95 % of all particles measuring less than 1 μm [101] and provided a stable reservoir of paclitaxel allowing for extended drug release. This was administered intraperitoneally in patients with solid tumors confined chiefly to the peritoneal cavity. All 21 patients had undergone chemotherapy and surgical procedures. Six doses of cremophor-free Nanotax (50–275 mg/m2) were administered in 2–2.5 L solutions for 30–60 min in one to six cycles, every 28 days. There were minimal side effects with regard to neutropenia or neurologic toxicities. One patient had thrombocytopenia that was unlikely related to Nanotax. The peritoneal concentrations were 450–2,900 times greater than the peak plasma concentrations and remained elevated through the entire dose cycle. Four patients were stable while 12 had increasing disease. Five patients survived greater than 400 days after initiation of the Nanotax treatment.
Mirza et al. [102] have just published the results of a double-blind phase 3 trial of Niraparib, an oral poly(adenosine diphosphate [ADP]-ribose) polymerase 1/2 inhibitor that has clinical activity in patients with ovarian cancer and which was proposed as maintenance therapy. In the cohort of 553 patients, there were both gBRCA and non-gBRCA cohorts that were assigned either Niraparib or placebo. Patients in the Niraparib group had a significantly longer median duration of progression-free survival (21 vs. 5.5 months in the gBRCA cohort and 9.3 vs. 3.9 months in the non-gBRCA cohort). Bone marrow toxicity resulted in 34 % thrombocytopenia, 25 % anemia and 20 % neutropenia, which were managed with dose modifications.
Conclusions
Significant progress in the treatment of peritoneal malignancies has been made over the last 20 years. Cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy has increased survival and extended lives. New agents and techniques of intraperitoneal therapy have been designed to insure coverage of the entire target area, penetration of the target tissue and in some cases targeting of the tumor with the goal of increasing survival times. However, most of these developments need more widespread clinical testing, and therefore, challenges still remain for researchers, oncologists, surgeons, and patients.
Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.
Research funding: None declared.
Employment or leadership: None declared.
Honorarium: None declared.
Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.
References
1. Roviello F, Roviello G, Petrioli R, Marrelli D. Hyperthermic intraperitoneal chemotherapy for the treatment of ovarian cancer: a brief overview of recent results. Crit Rev Oncol Hematol 2015;95:297–305.Search in Google Scholar
2. D’Angelica M, Gonen M, Brennan MF, Turnbull AD, Bains M, Karpeh MS. Patterns of initial recurrence in completely resected gastric adenocarcinoma. Ann Surg 2004;240:808–16.Search in Google Scholar
3. Winter JM, Tang LH, Klimstra DS, Liu W, Linkov I, Brennan MF, et al. Failure patterns in resected pancreas adenocarcinoma: lack of predicted benefit to SMAD4 expression. Ann Surg 2013;258:331–5.Search in Google Scholar
4. Sjo OH, Berg M, Merok MA, Kolberg M, Svindland A, Lothe RA, et al. Peritoneal carcinomatosis of colon cancer origin: highest incidence in women and in patients with right-sided tumors. J Surg Oncol 2011;104:792–7.Search in Google Scholar
5. Mendenhall WM, Million RR, Pfaff WW. Patterns of recurrence in adenocarcinoma of the rectum and rectosigmoid treated with surgery alone: implications in treatment planning with adjuvant radiation therapy. Int J Radiat Oncol Biol Phys 1983;9:977–85.Search in Google Scholar
6. Sugarbaker PH. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the management of gastrointestinal cancers with peritoneal metastases: progress toward a new standard of care. Cancer Treat Rev 2016;48:42–9.Search in Google Scholar
7. Chua TC, Robertson G, Liauw W, Farrell R, Yan TD, Morris DL. Intraoperative hyperthermic intraperitoneal chemotherapy after cytoreductive surgery in ovarian cancer peritoneal carcinomatosis: systematic review of current results. J Cancer Res Clin Oncol 2009;135:1637–45.Search in Google Scholar
8. Girshally R, Demtroder C, Albayrak N, Zieren J, Tempfer C, Reymond MA. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) as a neoadjuvant therapy before cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. World J Surg Oncol 2016;14:253.Search in Google Scholar
9. Dedrick RL, Flessner MF. Pharmacokinetic problems in peritoneal drug administration: tissue penetration and surface exposure. J Natl Cancer Inst 1997;89:480–7.Search in Google Scholar
10. Flessner MF. Computerized kinetic modeling: a new tool in the quest for adequacy in peritoneal dialysis. Perit Dial Int 1997;17:581–5.Search in Google Scholar
11. Flessner MF. Peritoneal transport physiology: insights from basic research. J Am Soc Nephrol 1991;2:122–35.Search in Google Scholar
12. Flessner MF, Fenstermacher JD, Dedrick RL, Blasberg RG. A distributed model of peritoneal-plasma transport: tissue concentration gradients. Am J Physiol 1985;248:F425–35.Search in Google Scholar
13. Lu Z, Wang J, Wientjes MG, Au JL. Intraperitoneal therapy for peritoneal cancer. Future Oncol 2010;6:1625–41.Search in Google Scholar
14. Collins JM, Dedrick RL, Flessner MF, Guarino AM. Concentration-dependent disappearance of fluorouracil from peritoneal fluid in the rat: experimental observations and distributed modeling. J Pharm Sci 1982;71:735–8.Search in Google Scholar
15. Los G, Mutsaers PH, van der Vijgh WJ, Baldew GS, de Graaf PW, McVie JG. Direct diffusion of cis-diamminedichloroplatinum(II) in intraperitoneal rat tumors after intraperitoneal chemotherapy: a comparison with systemic chemotherapy. Cancer Res 1989;49:3380–4.Search in Google Scholar
16. Los G, Mutsaers PH, Lenglet WJ, Baldew GS, McVie JG. Platinum distribution in intraperitoneal tumors after intraperitoneal cisplatin treatment. Cancer Chemother Pharmacol 1990;25:389–94.Search in Google Scholar
17. Goodman MD, McPartland S, Detelich D, Saif MW. Chemotherapy for intraperitoneal use: a review of hyperthermic intraperitoneal chemotherapy and early post-operative intraperitoneal chemotherapy. J Gastrointest Oncol 2016;7:45–57.Search in Google Scholar
18. Au JL, Guo P, Gao Y, Lu Z, Wientjes MG, Tsai M. Multiscale tumor spatiokinetic model for intraperitoneal therapy. AAPS J 2014;16:424–39.Search in Google Scholar
19. Del Vecchio S, Reynolds JC, Blasberg RG, Neumann RD, Carrasquillo JA, Hellstrom I, et al. Measurement of local Mr 97,000 and 250,000 protein antigen concentration in sections of human melanoma tumor using in vitro quantitative autoradiography. Cancer Res 1988;48:5475–81.Search in Google Scholar
20. Fujimori K, Covell DG, Fletcher JE, Weinstein JN. A modeling analysis of monoclonal antibody percolation through tumors: a binding-site barrier. J Nucl Med 1990;31:1191–8.Search in Google Scholar
21. Fujimori K, Covell DG, Fletcher JE, Weinstein JN. Modeling analysis of the global and microscopic distribution of immunoglobulin G, F(ab’)2, and Fab in tumors. Cancer Res 1989;49:5656–63.Search in Google Scholar
22. Dower SK, Ozato K, Segal DM. The interaction of monoclonal antibodies with MHC class I antigens on mouse spleen cells. I. Analysis of the mechanism of binding. J Immunol 1984;132:751–8.Search in Google Scholar
23. Averbach AM, Sugarbaker PH. Methodologic considerations in treatment using intraperitoneal chemotherapy. Cancer Treat Res 1996;82:289–309.Search in Google Scholar
24. Sugarbaker PH. Management of peritoneal-surface malignancy: the surgeon’s role. Langenbecks Arch Surg 1999;384:576–87.Search in Google Scholar
25. Yonemura Y, Fujimura T, Fushida S, Takegawa S, Kamata T, Katayama K, et al. Hyperthermo-chemotherapy combined with cytoreductive surgery for the treatment of gastric cancer with peritoneal dissemination. World J Surg 1991;15:530–5; discussion 5–6.Search in Google Scholar
26. Fujimura T, Yonemura Y, Muraoka K, Takamura H, Hirono Y, Sahara H, et al. Continuous hyperthermic peritoneal perfusion for the prevention of peritoneal recurrence of gastric cancer: randomized controlled study. World J Surg 1994;18:150–5.Search in Google Scholar
27. Elias D, Dube P, Blot F, Bonvallot S, Eggenspieler P, Sabourin JC, et al. Peritoneal carcinomatosis treatment with curative intent: the Institut Gustave-Roussy experience. Eur J Surg Oncol 1997;23:317–21.Search in Google Scholar
28. Fujimoto S, Shrestha RD, Kokubun M, Ohta M, Takahashi M, Kobayashi K, et al. Intraperitoneal hyperthermic perfusion combined with surgery effective for gastric cancer patients with peritoneal seeding. Ann Surg 1988;208:36–41.Search in Google Scholar
29. Koga S, Hamazoe R, Maeta M, Shimizu N, Murakami A, Wakatsuki T. Prophylactic therapy for peritoneal recurrence of gastric cancer by continuous hyperthermic peritoneal perfusion with mitomycin C. Cancer 1988;1561:232–7.Search in Google Scholar
30. Lotti M, Capponi MG, Piazzalunga D, Poiasina E, Pisano M, Manfredi R, et al. Laparoscopic HIPEC: a bridge between open and closed-techniques. J Minim Access Surg 2016;12:86–9.Search in Google Scholar
31. Spratt JS, Adcock RA, Sherrill W, Travathen S. Hyperthermic peritoneal perfusion system in canines. Cancer Res 1980;40:253–5.Search in Google Scholar
32. Spratt JS, Adcock RA, Muskovin M, Sherrill W, McKeown J. Clinical delivery system for intraperitoneal hyperthermic chemotherapy. Cancer Res 1980;40:256–60.Search in Google Scholar
33. Hildebrandt B, Wust P, Ahlers O, Dieing A, Sreenivasa G, Kerner T, et al. The cellular and molecular basis of hyperthermia. Crit Rev Oncol Hematol 2002;43:33–56.Search in Google Scholar
34. Issels RD. Regional hyperthermia in high-risk soft tissue sarcomas. Curr Opin Oncol 2008;20:438–43.Search in Google Scholar
35. Kampinga HH. Hyperthermia, thermotolerance and topoisomerase II inhibitors. Br J Cancer 1995;72:333–8.Search in Google Scholar
36. de Bree E, Witkamp AJ, Zoetmulder FA. Peroperative hyperthermic intraperitoneal chemotherapy (HIPEC) for advanced gastric cancer. Eur J Surg Oncol 2000;26:630–2.Search in Google Scholar
37. Witkamp AJ, de Bree E, Van Goethem R, Zoetmulder FA. Rationale and techniques of intra-operative hyperthermic intraperitoneal chemotherapy. Cancer Treat Rev 2001;27:365–74.Search in Google Scholar
38. Witkamp AJ, de Bree E, Kaag MM, Boot H, Beijnen JH, van Slooten GW, et al. Extensive cytoreductive surgery followed by intra-operative hyperthermic intraperitoneal chemotherapy with mitomycin-C in patients with peritoneal carcinomatosis of colorectal origin. Eur J Cancer 2001;37:979–84.Search in Google Scholar
39. Witkamp AJ, de Bree E, Kaag MM, van Slooten GW, van Coevorden F, Zoetmulder FA. Extensive surgical cytoreduction and intraoperative hyperthermic intraperitoneal chemotherapy in patients with pseudomyxoma peritonei. Br J Surg 2001;88:458–63.Search in Google Scholar
40. de Bree E, Witkamp AJ, Zoetmulder FA. Hyperthermic intraperitoneal chemoperfusion in the treatment of locally advanced intra-abdominal cancer. Br J Surg 2001;88:152.Search in Google Scholar
41. Zakaria ER, Lofthouse J, Flessner MF. Effect of intraperitoneal pressures on tissue water of the abdominal muscle. Am J Physiol Renal Physiol 2000;278:F875–85.Search in Google Scholar
42. Flessner MF. Small-solute transport across specific peritoneal tissue surfaces in the rat. J Am Soc Nephrol 1996;7:225–33.Search in Google Scholar
43. Flessner MF, Dedrick RL, Schultz JS. A distributed model of peritoneal-plasma transport: analysis of experimental data in the rat. Am J Physiol 1985;248:F413–24.Search in Google Scholar
44. Flessner MF, Dedrick RL, Reynolds JC. Bidirectional peritoneal transport of immunoglobulin in rats: tissue concentration profiles. Am J Physiol 1992;263:F15–23.Search in Google Scholar
45. Flessner MF, Dedrick RL, Reynolds JC. Bidirectional peritoneal transport of immunoglobulin in rats: compartmental kinetics. Am J Physiol 1992;262:F275–87.Search in Google Scholar
46. Flessner MF, Dedrick RL. Tissue-level transport mechanisms of intraperitoneally-administered monoclonal antibodies. J Control Release 1998;53:69–75.Search in Google Scholar
47. Flessner MF. The transport barrier in intraperitoneal therapy. Am J Physiol Renal Physiol 2005;288:F433–42.Search in Google Scholar
48. Flessner MF, Choi J, Credit K, Deverkadra R, Henderson K. Resistance of tumor interstitial pressure to the penetration of intraperitoneally delivered antibodies into metastatic ovarian tumors. Clin Cancer Res 2005;11:3117–25.Search in Google Scholar
49. Choi J, Credit K, Henderson K, Deverkadra R, He Z, Wiig H, et al. Intraperitoneal immunotherapy for metastatic ovarian carcinoma: resistance of intratumoral collagen to antibody penetration. Clin Cancer Res 2006;12:1906–12.Search in Google Scholar
50. Flessner MF. Intraperitoneal drug therapy: physical and and biological principles. Cancer Treat Res 2007;134:131–52.Search in Google Scholar
51. Jain RK. Barriers to drug delivery in solid tumors. Sci Am 1994;271:58–65.Search in Google Scholar
52. Boucher Y, Baxter LT, Jain RK. Interstitial pressure gradients in tissue-isolated and subcutaneous tumors: implications for therapy. Cancer Res 1990;150:4478–84.Search in Google Scholar
53. Less JR, Posner MC, Boucher Y, Borochovitz D, Wolmark N, Jain RK. Interstitial hypertension in human breast and colorectal tumors. Cancer Res 1992;52:6371–4.Search in Google Scholar
54. Boucher Y, Jain RK. Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse. Cancer Res 1992;52:5110–14.Search in Google Scholar
55. Roh HD, Boucher Y, Kalnicki S, Buchsbaum R, Bloomer WD, Jain RK. Interstitial hypertension in carcinoma of uterine cervix in patients: possible correlation with tumor oxygenation and radiation response. Cancer Res 1991;51:6695–8.Search in Google Scholar
56. Lu Z, Tsai M, Lu D, Wang J, Wientjes MG, Au JL. Tumor-penetrating microparticles for intraperitoneal therapy of ovarian cancer. J Pharmacol Exp Ther 2008;327:673–82.Search in Google Scholar
57. Gremonprez F, Descamps B, Izmer A, Vanhove C, Vanhaecke F, De Wever O, et al. Pretreatment with VEGF(R)-inhibitors reduces interstitial fluid pressure, increases intraperitoneal chemotherapy drug penetration, and impedes tumor growth in a mouse colorectal carcinomatosis model. Oncotarget 2015;6:29889–900.Search in Google Scholar
58. Reymond MA, Hu B, Garcia A, Reck T, Kockerling F, Hess J, et al. Feasibility of therapeutic pneumoperitoneum in a large animal model using a microvaporisator. Surg Endosc 2000;14:51–5.Search in Google Scholar
59. Solass W, Hetzel A, Nadiradze G, Sagynaliev E, Reymond MA. Description of a novel approach for intraperitoneal drug delivery and the related device. Surg Endosc 2012;26:1849–55.Search in Google Scholar
60. Solass W, Herbette A, Schwarz T, Hetzel A, Sun JS, Dutreix M, et al. Therapeutic approach of human peritoneal carcinomatosis with Dbait in combination with capnoperitoneum: proof of concept. Surg Endosc 2012;26:847–52.Search in Google Scholar
61. Tempfer CB, Rezniczek GA, Ende P, Solass W, Reymond MA. Pressurized intraperitoneal aerosol chemotherapy with cisplatin and doxorubicin in women with peritoneal carcinomatosis: a cohort study. Anticancer Res 2015;35:6723–9.Search in Google Scholar
62. Tempfer CB, Winnekendonk G, Solass W, Horvat R, Giger-Pabst U, Zieren J, et al. Pressurized intraperitoneal aerosol chemotherapy in women with recurrent ovarian cancer: a phase 2 study. Gynecol Oncol 2015;137:223–8.Search in Google Scholar
63. Tempfer CB. Pressurized intraperitoneal aerosol chemotherapy as an innovative approach to treat peritoneal carcinomatosis. Med Hypotheses 2015;85:480–4.Search in Google Scholar
64. Blanco A, Giger-Pabst U, Solass W, Zieren J, Reymond MA. Renal and hepatic toxicities after pressurized intraperitoneal aerosol chemotherapy (PIPAC). Ann Surg Oncol 2013;20:2311–16.Search in Google Scholar
65. Odendahl K, Solass W, Demtroder C, Giger-Pabst U, Zieren J, Tempfer C, et al. Quality of life of patients with end-stage peritoneal metastasis treated with pressurized intraperitoneal aerosol chemotherapy (PIPAC). Eur J Surg Oncol 2015;41:1379–85.Search in Google Scholar
66. Jung DH, Lee YC, Kim JH, Lee SK, Shin SK, Park JC, et al. Additive treatment improves survival in elderly patients after non-curative endoscopic resection for early gastric cancer. Surg Endosc 2016. [Epub ahead of print].Search in Google Scholar
67. Jung do H, Son SY, Oo AM, Park YS, Shin DJ, Ahn SH, et al. Feasibility of hyperthermic pressurized intraperitoneal aerosol chemotherapy in a porcine model. Surg Endosc 2016;30:4258–64.Search in Google Scholar
68. Kakchekeeva T, Demtroder C, Herath NI, Griffiths D, Torkington J, Solass W, et al. In vivo feasibility of electrostatic precipitation as an adjunct to pressurized intraperitoneal aerosol chemotherapy (ePIPAC). Ann Surg Oncol 2016. [Epub ahead of print].Search in Google Scholar
69. Solass W, Kerb R, Murdter T, Giger-Pabst U, Strumberg D, Tempfer C, et al. Intraperitoneal chemotherapy of peritoneal carcinomatosis using pressurized aerosol as an alternative to liquid solution: first evidence for efficacy. Ann Surg Oncol 2014;21:553–9.Search in Google Scholar
70. Khosrawipour V, Khosrawipour T, Falkenstein TA, Diaz-Carballo D, Forster E, Osma A, et al. Evaluating the effect of micropump(c) position, internal pressure and doxorubicin dosage on efficacy of Pressurized Intra-peritoneal Aerosol Chemotherapy (PIPAC) in an ex vivo model. Anticancer Res 2016;36:4595–600.Search in Google Scholar
71. Khosrawipour V, Khosrawipour T, Diaz-Carballo D, Forster E, Zieren J, Giger-Pabst U. Exploring the spatial drug distribution pattern of pressurized intraperitoneal aerosol chemotherapy (PIPAC). Ann Surg Oncol 2016;23:1220–4.Search in Google Scholar
72. Khosrawipour V, Giger-Pabst U, Khosrawipour T, Pour YH, Diaz-Carballo D, Forster E, et al. Effect of irradiation on tissue penetration depth of doxorubicin after Pressurized Intra-Peritoneal Aerosol Chemotherapy (PIPAC) in a novel ex-vivo model. J Cancer 2016;7:910–14.Search in Google Scholar
73. Khosrawipour V, Khosrawipour T, Kern AJ, Osma A, Kabakci B, Diaz-Carballo D, et al. Distribution pattern and penetration depth of doxorubicin after pressurized intraperitoneal aerosol chemotherapy (PIPAC) in a postmortem swine model. J Cancer Res Clin Oncol 2016;142:2275–80.Search in Google Scholar
74. Flessner MF, Fenstermacher JD, Blasberg RG, Dedrick RL. Peritoneal absorption of macromolecules studied by quantitative autoradiography. Am J Physiol 1985;248:H26–32.Search in Google Scholar
75. Hirano K, Hunt CA. Lymphatic transport of liposome-encapsulated agents: effects of liposome size following intraperitoneal administration. J Pharm Sci 1985;74:915–21.Search in Google Scholar
76. Armstrong DK, Fleming GF, Markman M, Bailey HH. A phase I trial of intraperitoneal sustained-release paclitaxel microspheres (Paclimer) in recurrent ovarian cancer: a Gynecologic Oncology Group study. Gynecol Oncol 2006;103:391–6.Search in Google Scholar
77. Shive MS, Anderson JM. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Deliv Rev 1997;28:5–24.Search in Google Scholar
78. Kuh HJ, Jang SH, Wientjes MG, Weaver JR, Au JL. Determinants of paclitaxel penetration and accumulation in human solid tumor. J Pharmacol Exp Ther 1999;290:871–80.Search in Google Scholar
79. Jang SH, Wientjes MG, Au JL. Enhancement of paclitaxel delivery to solid tumors by apoptosis-inducing pretreatment: effect of treatment schedule. J Pharmacol Exp Ther 2001;296:1035–42.Search in Google Scholar
80. Lu D, Wientjes MG, Lu Z, Au JL. Tumor priming enhances delivery and efficacy of nanomedicines. J Pharmacol Exp Ther 2007;322:80–8.Search in Google Scholar
81. Carlier C, Laforce B, Van Malderen SJ, Gremonprez F, Tucoulou R, Villanova J, et al. Nanoscopic tumor tissue distribution of platinum after intraperitoneal administration in a xenograft model of ovarian cancer. J Pharm Biomed Anal 2016;30:256–62.Search in Google Scholar
82. Tsai M, Lu Z, Wang J, Yeh TK, Wientjes MG, Au JL. Effects of carrier on disposition and antitumor activity of intraperitoneal paclitaxel. Pharm Res 2007;24:1691–701.Search in Google Scholar
83. Tsai M, Lu Z, Wientjes MG, Au JL. Paclitaxel-loaded polymeric microparticles: quantitative relationships between in vitro drug release rate and in vivo pharmacodynamics. J Control Release 2013;172:737–44.Search in Google Scholar
84. Au JL, Lu Z, Wientjes MG. Versatility of particulate carriers: development of pharmacodynamically optimized drug-loaded microparticles for treatment of peritoneal cancer. AAPS J 2015;17:1065–79.Search in Google Scholar
85. Di Pasqua AJ, Yuan H, Chung Y, Kim JK, Huckle JE, Li C, et al. Neutron-activatable holmium-containing mesoporous silica nanoparticles as a potential radionuclide therapeutic agent for ovarian cancer. J Nucl Med 2013;54:111–16.Search in Google Scholar
86. Fu Q, Hargrove D, Lu X. Improving paclitaxel pharmacokinetics by using tumor-specific mesoporous silica nanoparticles with intraperitoneal delivery. Nanomedicine 2016;12:1951–9.Search in Google Scholar
87. Ohta S, Hiramoto S, Amano Y, Sato M, Suzuki Y, Shinohara M, et al. Production of cisplatin-incorporating hyaluronan nanogels via chelating ligand-metal coordination. Bioconjug Chem 2016;27:504–8.Search in Google Scholar
88. Teesalu T, Sugahara KN, Ruoslahti E. Tumor-penetrating peptides. Front Oncol 2013;3:216.Search in Google Scholar
89. Teesalu T, Sugahara KN, Kotamraju VR, Ruoslahti E. C-end rule peptides mediate neuropilin-1-dependent cell, vascular, and tissue penetration. Proc Natl Acad Sci U S A 2009;106:16157–62.Search in Google Scholar
90. Sugahara KN, Scodeller P, Braun GB, de Mendoza TH, Yamazaki CM, Kluger MD, et al. A tumor-penetrating peptide enhances circulation-independent targeting of peritoneal carcinomatosis. J Control Release 2015;28:59–69.Search in Google Scholar
91. Sugahara KN, Braun GB, de Mendoza TH, Kotamraju VR, French RP, Lowy AM, et al. Tumor-penetrating iRGD peptide inhibits metastasis. Mol Cancer Ther 2015;14:120–8.Search in Google Scholar
92. Sugahara KN, Teesalu T, Karmali PP, Kotamraju VR, Agemy L, Greenwald DR, et al. Coadministration of a tumor-penetrating peptide enhances the efficacy of cancer drugs. Science 2010;328:1031–5.Search in Google Scholar
93. Simon-Gracia L, Hunt H, Scodeller P, Gaitzsch J, Kotamraju VR, Sugahara KN, et al. iRGD peptide conjugation potentiates intraperitoneal tumor delivery of paclitaxel with polymersomes. Biomaterials 2016;104:247–57.Search in Google Scholar
94. Simon-Gracia L, Hunt H, Scodeller PD, Gaitzsch J, Braun GB, Willmore AM, et al. Paclitaxel-loaded polymersomes for enhanced intraperitoneal chemotherapy. Mol Cancer Ther 2016;15:670–9.Search in Google Scholar
95. Fagotti A, Paris I, Grimolizzi F, Fanfani F, Vizzielli G, Naldini A, et al. Secondary cytoreduction plus oxaliplatin-based HIPEC in platinum-sensitive recurrent ovarian cancer patients: a pilot study. Gynecol Oncol 2009;113:335–40.Search in Google Scholar
96. Fagotti A, Costantini B, Vizzielli G, Perelli F, Ercoli A, Gallotta V, et al. HIPEC in recurrent ovarian cancer patients: morbidity-related treatment and long-term analysis of clinical outcome. Gynecol Oncol 2011;122:221–5.Search in Google Scholar
97. Fagotti A, Costantini B, Petrillo M, Vizzielli G, Fanfani F, Margariti PA, et al. Cytoreductive surgery plus HIPEC in platinum-sensitive recurrent ovarian cancer patients: a case-control study on survival in patients with two year follow-up. Gynecol Oncol 2012;127:502–5.Search in Google Scholar
98. Petrillo M, De Iaco P, Cianci S, Perrone M, Costantini B, Ronsini C, et al. Long-term survival for platinum-sensitive recurrent ovarian cancer patients treated with secondary cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy (HIPEC). Ann Surg Oncol 2016;23:1660–5.Search in Google Scholar
99. Fagotti A, Ferrandina G, Vizzielli G, Fanfani F, Gallotta V, Chiantera V, et al. Phase III randomised clinical trial comparing primary surgery versus neoadjuvant chemotherapy in advanced epithelial ovarian cancer with high tumour load (SCORPION trial): final analysis of peri-operative outcome. Eur J Cancer 2016;59:22–33.Search in Google Scholar
100. Williamson SK, Johnson GA, Maulhardt HA, Moore KM, McMeekin DS, Schulz TK, et al. A phase I study of intraperitoneal nanoparticulate paclitaxel (Nanotax(R)) in patients with peritoneal malignancies. Cancer Chemother Pharmacol 2015;75:1075–87.Search in Google Scholar
101. Roby KF, Niu F, Rajewski RA, Decedue C, Subramaniam B, Terranova PF. Syngeneic mouse model of epithelial ovarian cancer: effects of nanoparticulate paclitaxel, Nanotax. Adv Exp Med Biol 2008;622:169–81.Search in Google Scholar
102. Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med 2016. [Epub ahead of print].Search in Google Scholar
©2016 by De Gruyter Mouton
Articles in the same Issue
- Frontmatter
- Editorial
- The quest of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC): searching for evidence
- Reviews
- Randomized controlled trials evaluating cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) in prevention and therapy of peritoneal metastasis: a systematic review
- Pharmacokinetic problems in peritoneal drug administration: an update after 20 years
- Opinion Paper
- Morphology of the peritoneal cavity and pathophysiological consequences
- Research Articles
- Environmental safety during the administration of Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC)
- Preoperative intraperitoneal oxaliplatin for unresectable peritoneal carcinomatosis of colorectal origin: a pilot study
- Single-port access for Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC): technique, feasibility and safety
Articles in the same Issue
- Frontmatter
- Editorial
- The quest of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC): searching for evidence
- Reviews
- Randomized controlled trials evaluating cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) in prevention and therapy of peritoneal metastasis: a systematic review
- Pharmacokinetic problems in peritoneal drug administration: an update after 20 years
- Opinion Paper
- Morphology of the peritoneal cavity and pathophysiological consequences
- Research Articles
- Environmental safety during the administration of Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC)
- Preoperative intraperitoneal oxaliplatin for unresectable peritoneal carcinomatosis of colorectal origin: a pilot study
- Single-port access for Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC): technique, feasibility and safety