Intraperitoneal Drug Delivery for Ovarian Cancer: Why, How, Who, What, and When?

Publication
Article
OncologyONCOLOGY Vol 25 No 2
Volume 25
Issue 2

Epithelial ovarian cancer (EOC) spreads prominently within the peritoneal cavity. In fact, we now know that high-grade serous cancers are often of tubal origin, and their presentation as tubo-ovarian masses renders it likely that intraperitoneal spread occurs as an early event in their clinical evolution.

In 1996, intraperitoneal (IP) administration of cisplatin plus intravenous (IV) cyclophosphamide proved superior to both drugs given intravenously at the same doses-which, at the time, was the standard treatment in the United States. The IP ‘option’ was not adopted, however, because the standard treatment had shifted to IV cisplatin plus paclitaxel. Two additional phase III trials by the Gynecologic Oncology Group (GOG) comparing IP versus IV cisplatin, but including other variables, have shown similar superior effects of the IP route on outcome, but with toxicities-particularly local tolerance and neuropathy-increased. An ongoing trial by the GOG is again looking into an IP versus IV comparison, and introducing in one of the IP arms the substitution of IP carboplatin for IP cisplatin. All three arms of this trial contain bevacizumab (Avastin). Two other trials comparing IV versus IP administration of platinums or platinums and paclitaxel have just been launched, led by Japanese and Canadian investigators, respectively. While awaiting additional data on the ongoing debate over IP versus IV therapy, it is important that we consider issues concerning why the IP route may be relevant, how can one increase the safety of this route, and who should be treated and with what drugs, particularly when faced with a patient outside the clinical trials setting. The underlying hypothesis for use of IP therapy is based on the existence of a dose-effect relationship for platinum drugs in ovarian cancer. We review the known data on this relationship, and explore why interest in platinum drugs has become the central focus of ovarian cancer treatment.

Epithelial ovarian cancer (EOC) spreads prominently within the peritoneal cavity. In fact, we now know that high-grade serous cancers are often of tubal origin, and their presentation as tubo-ovarian masses renders it likely that intraperitoneal spread occurs as an early event in their clinical evolution. From the outset, IP drug administration in ovarian cancer conferred a pharmacologic advantage over IV administration, given the additional clinical benefit derived from achieving more efficient control of life-threatening peritoneal disease.[1]

The systemic treatment of epithelial ovarian cancer and related extrauterine adenocarcinomas of Mullerian origin relies on ‘platinum-based’ chemotherapy. Ever since the introduction of cisplatin into our armamentarium in the late 1970s,[2] this drug, and later its less toxic analog, carboplatin, have provided the platform for treatment of these malignancies at all stages. Adoption of cisplatin as standard treatment accelerated with the discovery of effective antiemetics, and introduction of the equally efficacious carboplatin, with its more predictable pharmacokinetics and superior safety profile, led to wide exploration of dose-response relationships.[3,4] Before meta-analyses were in vogue, William Hryniuk and colleagues observed that in ovarian cancer trials, the dose of cisplatin correlated with improved survival.[5] However, evidence for benefit beyond a certain ‘dose-intensity’ was not obvious.[6] As Martin Gore noted in a 2003 editorial in the Journal of Clinical Oncology, “Devotees of the ‘more is better’ school of oncology must face the uncomfortable truth that the majority of randomized trials have failed to show an overall survival benefit associated with an increase in the total dose or dose-intensity of platinum.” He went on to state, however, “Any discussion of dose-response in ovarian cancer is incomplete without a reference to the subject that always guarantees a thoroughly entertaining argument: namely, intraperitoneal chemotherapy. The data from randomized trials of intraperitoneal chemotherapy do support the premise that a dose-response exists....”[7]

TABLE 1


Results of the Three GOG Phase III Trials and Key Criticisms Against Their Message

This discussion is still applicable from today’s perspective, but it has become even more relevant than when Gore’s editorial was written. In 2006 the US National Cancer Institute (NCI) issued a clinical announcement[8] after the results of the third of phase III GOG trials (GOG172) on IP versus IV became known and were subsequently published (by Armstrong, in the New England Journal of Medicine, in 1996, and others).[9-11] After reviewing the data from the IP versus IV phase III clinical trials conducted by the GOG (Table 1), the NCI announced, “Based on the results of these randomized phase III trials, a combination of IV and IP administration of chemotherapy conveys a significant survival benefit among women with optimally debulked epithelial ovarian cancer, compared to IV administration alone.” Further, the NCI urged physicians to become familiar with these trials and, if necessary, refer patients to centers familiar with IP drug delivery. Both in the US and Europe, reaction to such announcement was swift: Ozols and colleagues noted that “…exploratory cross-trial comparisons of IV carboplatin/paclitaxel compared to IP regimens suggest very similar efficacy in optimal stage III disease,”[12] and Gore et al argued, ‘There are at least eight reasons why the Armstrong article does not support the use of IP administration as a standard of care.”[13]

FIGURE 1

Schema and outcome of GOG172. GOG = Gynecologic Oncology Group; SWOG = Southwest Oncology Group. Information from Reference 11.FIGURE 2

GOG172 debated areas. (Figure is a adapted from Nicoletta Colombo presentation at the 10th International Conference on Platinum Coordination Compounds in Cancer Chemotherapy, Verona, Italy, 2007.) GOG = Gynecologic Oncology Group.

A major problem in the adoption of IP therapy as the new standard for optimally debulked ovarian cancer might be put forth in a format analogous to the title of Gore’s 2003 editorial in JCO: “IP Cisplatin Is More Effective than IV Cisplatin: But How Do I Prescribe It?”[7] Figure 1 shows the schema of GOG172 and Figure 2 graphically lays out many of the disputed areas in GOG172, including: Is IP paclitaxel necessary to achieve the results? Is the dose of IP cisplatin of 100 mg/m2 acceptable to many patients? How does one deal with the 24-hour infusions of paclitaxel? If one had used a carboplatin control, would the survival differences have been significant? Can one advocate a treatment with a 42% completion rate? And, is the tradeoff of adverse quality of life for a few months and risk of treatment complications from IP therapy for a less than 20% reduction in risk of dying reasonable? GOG investigators struggled with these issues, and after a series of pilot studies embarked in late 2009 on another IP versus IV phase III study (GOG252) containing also an IP toxicity–reduction arm, based on IP carboplatin and phase II data from Japan.[14]

With this as a background, we expand on this debate and provide some additional perspective: the effect of platinum drugs on the outcome of ovarian cancer is even more relevant today than it was in the past, given the apparent shortcomings of many new drugs that have been tested. The route of platinum administration, as well as patient selection and the role of other drugs, should be central to ongoing research in this area.

How? An Outline of Methods for IP Drug Delivery

IP drug delivery is an old topic. The fact that the administration of IP chemotherapy is more complex and time-consuming than IV chemotherapy, has been associated with a greater risk of toxicity, and is both physically and emotionally more challenging for patients continues to shape the nonacceptance of this modality by clinicians. There is also a learning-curve in acquiring the proper techniques for surgically placing an IP port and developing the clinical expertise in accessing the IP port for drug delivery. There continue to be debates beyond the scope of our expertise regarding whether catheters with multiple holes or a single one should be used and the location of their placement. Also, insistence on heating solutions and making patients roll during the infusion seems misplaced. GOG institutions continue to accrue in a satisfactory way to the current trial, as they did in the past. GOG172 did have a greater problem in completion rate relative to the two prior GOG phase III trials, and also a greater number of catheter problems.[11,15,16] This higher complication rate in the last trial has been attributed to the greater aggressiveness of the debulking surgery performed, but is likely also due to the added IP access (twice every 3 weeks) and local toxicity that occurs with addition of IP paclitaxel.

From the extensive experience in past studies, some observations on use of the IP route included the following:

• Multiple surgeries increase the incidence of catheter malfunction;

• Certain vesicant drugs (eg, mitoxantrone) increase catheter malfunction[17,18];

FIGURE 3

IP contrast computerized tomography study showing optimal distribution of drug to peritoneal surfaces.

• Drug delivery is generally more satisfactory in patients selected for IP therapy based on low-volume disease[19] (Figure 3);

• Repeated percutaneous drug administration, while feasible, does not have a satisfactory record of completion (example: GONO trial)[20];

• At least for recurrent disease, volumes greater than 1 cm do not respond to any extent to IP drug administration (and the rationale for greater delivery of IP drug over IV administered drug to those tumor nodules is weak)[21]; and

• Single institutions with sufficient experience in IP drug delivery have a low incidence of complications from IP drug administration.[22]

While these ‘dogmas’ should be appropriately challenged by new data, they should still serve as general guidelines for satisfactory IP drug delivery.

From years of experience, certain procedural details emerge as important. Nurses experienced in the administration of IP chemotherapy are essential; they have the technical expertise in managing the “mechanics” of IP drug delivery and can meet the challenge of helping patients cope with the physical and emotional demands of undergoing IP therapy. Any problem with gravity administration of IP chemotherapy is a signal to reinspect whether the IP catheter is patent. If it is uncertain, rather than wasting precious time in clinical facilities, it is best to have a radiologist determine whether the catheter placement is correct and uncover any reasons for the inflow obstruction (usually loculation or complete occlusion, causing backflow around the cather and into the subcutaneous tissues). Regional side effects such as pain and bloating from abdominal distension are common problems with the initial delivery of chemotherapy via the peritoneal route. All women do not tolerate the same amount of IP fluid volume: a rough guideline we have adopted is using 1 L/m2.

Outflow obstruction is common: it is probably related to a small amount of connective tissue within the rim of the outflow orifice. In phase II/pharmacologic studies, not infrequently we collect specimens (including cytology) if outflow is satisfactory. The longer an IP catheter is left in place, however, the percentage of outflow obstruction rises to above 40%. If it is desirable, it is best to maintain the patency of the IP catheter by flushing the system with heparinized saline every 6 weeks. Nowadays, IV bevacizumab is often coadministered and continued beyond IP therapy,[23] raising the new question of when is it most desirable to remove the port and catheter under such circumstances. We did observe an episode of chemical peritonitis, when IV pegylated liposomal doxorubicin was continued in a patient who had not undergone removal of a previously placed IP catheter (FM, personal observation).

Who Has Been Shown to Benefit From IP Therapy?

Single-institution reports of long-term survivors[1] following failures of IV drug administration provided initial support for IP drug administration[24-31] and eventually spurred proof-of-concept randomized trials. These initial IP phase I/II and pharmacologic studies were reported throughout the 1980s, suggesting that survival was prolonged in patients who received salvage IP therapy after initial chemotherapy and second-look surgery (with minimal residual disease). Eventually, randomized studies provided the key evidence for efficacy applicable to patients diagnosed with high-grade serous epithelial ovarian cancer (and those of tubal and primary peritoneal origin) that predominantly spread to the omentum and the peritoneal cavity by intraabdominal dissemination (thus, stage III), and also to pelvic and paraaortic nodes by lymphatic drainage (locoregional disease).

TABLE 2


IP Versus IV Therapy in Ovarian Cancer: Non-GOG Studies

Eight randomized controlled trials have assessed IP therapy versus IV chemotherapy for first-line treatment in EOC. While the three studies performed by GOG (Table 1) encountered substantial criticisms, they were adequately powered and the differences shown are persuasively linked to the assigned route of administration. In the five trials shown in Table 2, there were no statistically significant differences, but the studies were uninterpretable because of lack of statistical power, in some clearly attributed to their early termination. The inability to adequately conduct these studies is a reflection of some of the inherent difficulties faced by clinical investigators involved in testing IP therapy.

After the NCI announcement, many groups in the US adopted GOG172 or its modifications for stage III optimally cytoreduced ovarian cancer. In the subsequent years, use of such IP therapy outside a trial setting may actually have declined, mainly because of difficult patient acceptance and competing clinical study options. For US GOG institutions, decisions have now been simplified because of availability of GOG252 as a treatment choice, until 2013. Who should be receiving IP therapy beyond the confines of a clinical trial? And paraphrasing Martin Gore, how do we prescribe it? Some historical perspective is helpful: when IP therapy became widely applied in the 1980s within the US, a number of phase II studies disclosed results focusing primarily on response rates determined by surgical reassessment-an inappropriate endpoint for patients who have primarily low-volume disease.

The GOG, in fact, opened a series of drug studies (GOG102 series) that were mostly inconclusive, except for finding that platinum sensitivity was a determinant of outcome.[32] Another determinant of outcome was the volume of disease (bulk disease may, in fact, correlate to some extent with the absence of platinum sensitivity). After years of uncontrolled series within institutions and cooperative groups, Robert Ozols in 1991 wrote an editorial entitled “IP Therapy of Ovarian Cancer: Time’s Up,”[33] and certainly it was about time to move on from these types of trials. Fortunately, since June 1986, the Southwest Oncology Group (SWOG) under David Alberts’ leadership had started an IP versus IV trial in the population most likely to be sensitive to cisplatin: previously untreated patients. It later became an intergroup study (GOG104), with GOG enrolling 295 patients, 298 enrolled by SWOG, and 61 by the Eastern Cooperative Oncology Group. In January 1991, with no knowledge of outcome, an amendment was passed to expand the study by 1 year “to achieve a large enough sample for a separate analysis of data from patients with residual tumors that were no larger than 0.5 cm.” Such an amendment was later a source of much criticism (see below). The study completed accrual in July 1992 and was published in December 1996.[9]

TABLE 3


Summary of Studies With Intraperitoneal Drugs

This first phase III trial showed an advantage for progression-free survival (PFS) and overall survival (OS) for the IP arm. Criticisms would follow this trial and would further escalate upon publication of the two subsequent phase III trials, also summarized in Table 1. While many of the criticisms are valid, including the multiple variables introduced into the IP versus IV question in subsequent trials, the common thread in the superior results is the IP administration of cisplatin. The three GOG trials taken together lead to the following perspective: 1) the IP route for cisplatin was favored in all three trials; 2) the results were achieved in previously untreated patients, when they would be expected to be most sensitive to platinums; and 3) IV carboplatin, while outperforming cisplatin (nonsignificantly) in some trials, is unlikely to yield significantly superior results over IV cisplatin (and why would one expect ovarian cancer to be different from any other neoplasm in this regard?). With respect to the greater toxicity of IP therapy, two factors are obvious contributors: 1) the dose of IP cisplatin being higher than the IV dose in the last two trials is a major contributor to the greater intolerance and would be expected to improve by a decrease to 75 mg/m2 (well documented in IV studies); and 2) IP paclitaxel, by accessing the peritoneal cavity twice as often, carries higher toxicity, and the drug itself adds to both local and systemic toxicities (especially neuropathy). Diminishing the dose of cisplatin has widely been adopted; however, eliminating IP paclitaxel meets with resistance by ‘pure’ trialists. Given that two studies omitting IP paclitaxel led to efficacy similar to that seen in the third trial, in our view superiority of IP paclitaxel regimens to either cisplatin or carboplatin alone given by the IP route must be demonstrated before there can be wider adoption of the GOG172 modified regimen.

IP treatment, nevertheless, should be considered for patients of similar stage and residuum who had been included in the three GOG trials. GOG104 included patients with < 2 cm residual disease-a somewhat more liberal entry criterion, explaining in part the lesser median survival time observed relative to the later trials. However, significant benefit of IP over IV was not demonstrated in the subset with smallest volumes of disease, indicating that the patients with larger residuum contributed to the outcome. This could argue in favor of not needing to be so strict with optimal cytoreduction-a topic we take up later in this article. It is noteworthy that GOG114 has the best PFS result of all the three trials, and provocatively this could lead to the interpretation that ‘chemical cytoreduction’ by IV carboplatin has rendered the efficacy of IP cisplatin more demonstrable. At present, one would continue to consider the best candidates for IP therapy those who are previously untreated and optimally cytoreduced.

Finally, what if one introduces a biomarker of platinum sensitivity to the selection of patients for IP cisplatin? Krivak and coworkers have used BRCA1 protein immunostaining as a surrogate for platinum sensitivity (with low BRCA1 expression occurring in about 50% of patients entered into GOG172, being considered as indicative of greater sensitivity to platinum).[34] In their retrospective study, 93 patients with low BRCA1 expression who received IV treatment were compared with 96 patients, also with low BRCA1 expression, who received IP treatment. Their median PFS times were 18.7 and 34.7 months, respectively, and their median OS times were 47.7 and 84.1 months, respectively. There were no differences in PFS or OS for patients treated by the IV versus IP method in the BRCA1 high-immunostaining patients. Certainly, if validated, this biomarker could greatly enhance the applicability of IP therapy by leading to the proper selection of patients. Also, the results are noteworthy on theoretical grounds: if the most sensitive patients benefit, this tells us that it is the platinum dose-intensity achieved by the IP route and not the relatively small increments achievable via the IV route that determine outcome. Conversely, even a greater than 10-fold increase in platinum exposure may be futile if platinum resistance is present.

What Drugs Should One Use?

The underlying principles for effective IP therapy have been most clearly articulated by Dedrick and coworkers when introducing it into clinical use,[35] and by Howell in molding its current clinical application. The following factors are taken into account.

Peritoneum Features

The peritoneal membrane has been used for many years for peritoneal dialysis, as a semipermeable membrane allowing exchange between the peritoneal cavity and the mesenteric blood vessels. Depending on the molecular weight, size, and the water solubility, larger-molecular-weight drugs stay longer within the peritoneal cavity while smaller molecules may penetrate the outer cell layers of a tumor more easily, albeit diffusing outside the peritoneum faster. When IP chemotherapy is delivered, however, the drug reaches the tumor both by diffusion via the free tumor surface in the peritoneal cavity, and by capillary flow via the systemic circulation.

Pharmacologic Features

The pharmacological advantage of IP therapy relates to high intraperitoneal concentrations delivered to the tumor, with less systemic toxicity compared with IV administration, often expressed as the ratio between the AUC in the peritoneum and the AUC in plasma. The efficacy of IP therapy depends on the peritoneal/plasma concentration ratio (pharmacokinetics) of the different drugs, their systemic absorption, and the penetration into tumor tissue (limited to a depth of few millimeters [2-9 mm], under the tumor surface). Based on the phase III trials reviewed, the cytotoxic drugs widely used are cisplatin, carboplatin, and paclitaxel.

Cisplatin is rapidly and well absorbed systemically following IP administration, resulting in 50%–100% of systemic exposure compared with IV administration when comparable doses are given. Otherwise, peak IP concentrations are 10- to 20-fold compared with IV administration and provide very high IP concentrations for locoregional disease within a broad range of clinically effective systemic cisplatin dose-schedules (50–100 mg/m2) for disease outside the abdomen. Dose-limiting toxicity from IP administration is directly related to the systemic effects of cisplatin (myelosuppression, neurotoxicity).

Carboplatin is absorbed following IP administration 2–4 hours after infusion. Miyagi et al compared the pharmacokinetics of carboplatin in IP and IV spaces after IP or IV administration of carboplatin.[36] They demonstrated that 24-h platinum AUC in the serum was exactly the same regardless of IP of IV administration of carboplatin. However, the 24-h platinum AUC in the peritoneal cavity was approximately 17 times higher when carboplatin was administered by the IP route. They suggested that IP carboplatin was feasible as regional therapy and also as systemic chemotherapy. The GOG 158 trial (designed as a noninferiority study) demonstrated that the IV combination of carboplatin AUC 7.5 mg/mL/min plus paclitaxel 175 mg/m2 in a 3-hour infusion was not inferior to the IV combination of cisplatin 75 mg/m2 plus paclitaxel 135 mg/m2 in a 24-hour continuous infusion in terms of PFS and OS in patients with stage III EOC.[37] Because of these results IV carboplatin replaced IV cisplatin, but IP carboplatin has not been compared against cisplatin in the IP setting. One preclinical and one clinical study originally supported the superiority of IP cisplatin over IP carboplatin. The first was an animal model (rats with colon carcinoma) that showed cisplatin was more effective than carboplatin at penetrating into tumor nodules (10-fold more carboplatin than cisplatin was required to obtain comparable intratumoral platinum concentrations).[38] The second was an outdated retrospective study of 65 patients that showed a higher response rate in patients who received IP cisplatin at 100 mg/m2 than those who received IP carboplatin at 200–300 mg/m2.[39] A later retrospective analysis by Fujiwara et al on 165 EOC patients treated with IP carboplatin at doses of < 400 mg/m2 or ≥ 400 mg/m2 alone or in combination with cyclophosphamide or paclitaxel emphasized the importance of IP carboplatin dose-intensity on outcome.[40] The survival time was 24.5 months when the dose of carboplatin was < 400 mg/m2 and 51 months when the dose was ≥ 400 mg/m2. More recently another animal-model study compared the ability of IP cisplatin and IP carboplatin to penetrate into tumor nodules (2–9 mm) of nu/nu mice. Animals were treated with IP cisplatin at 10 mg/kg or equimolar dose of IP carboplatin at 12.4 mg/kg or an equitoxic dose of carboplatin at 85 mg/kg. The platinum tumor concentration was 3.4-fold higher in mice receiving cisplatin than in those receiving the equimolar dose of carboplatin; however, when cisplatin and carboplatin were given at equitoxic doses, the platinum tumor concentration was similar (and even nonsignificantly higher for carboplatin).[41] Based on these data, further clinical trials are reasonable to evaluate IP carboplatin efficacy.

Paclitaxel was deemed an attractive agent for IP administration in ovarian cancer based on the studies performed by the GOG. It has a high molecular weight and bulky structure, resulting in slow clearance from the peritoneal compartment, thus increasing the exposure of tumor in the peritoneal cavity. On the other hand, little is known about penetrance into tumors, and its tendency to form micelles may diminish its availability to tumor cell surfaces. It is metabolized in the liver, which increases the peritoneal/plasma concentration ratio. After IP paclitaxel infusion, the peak peritoneum/plasma concentration ratio is approximately 1,000, and this is maintained for 24–48 hours. Abdominal pain is the dose-limiting toxicity of IP paclitaxel at doses of more than 125 mg/m2.[42-44]

In the preceding section, clinical trials data supporting the use of IP therapy, and specifically IP cisplatin, were reviewed. From the standpoint of toxicity considerations, as well as the added complexity of treatment and the absence of validation beyond a single phase III trial, the case for using IP paclitaxel is less persuasive. The pragmatic approach of one of the authors (FM) in adopting GOG 114 (IV drug administration first, and no IP paclitaxel) as his guiding strategy is reflected in the following clinical vignette:

Clinical vignette 1-In June 2004, the patient, a 61-year-old woman, noted abdominal distension and underwent a workup, including computerized tomography (CT), that confirmed ascites and perihepatic as well as pelvic masses. The CA125 level was 185 U/mL on June 23. On July 20, TAH/BSO (total abdominal hysterectomy/bilateral salpingo-oophorectomy) appendectomy, pelvic lymph node dissection, and debulking were performed. The mass appeared to arise from the right fallopian tube; a cul-de-sac nodule, and tumor involving the surfaces of the diaphragm around the right lobe of the liver, were noted and resected. The right-diaphragm tumor measured 8 × 5.5 × 3.5 cm. The patient was optimally cytoreduced but did not have IP catheter placement. The recommendation in August 2004 was: “off protocol, I prefer early reassessment (at cycle 4) and then insertion of an IP catheter as long as the patient has a clinical complete response. The findings of a negative or positive reassessment then determine the number of cycles to be given as consolidation. She will consider this option in the next few days.” After 4 cycles with excellent tolerance, she underwent laparoscopic placement of an IP catheter. With negative findings, 2 cycles of IP cisplatin 60 mg/m2 were given. Subsequently, she tested positive for BRCA1 mutation. She is well 6 years later.

Therefore, in response to the question “What regimen should one use?,” one of us (FM) would answer the following: if an IP port has been placed and the patient fulfills other criteria for proceeding with IP therapy (no prior treatment, normal renal function), the patient should receive IV paclitaxel at 175 mg/m2 on day 1, followed by IP cisplatin at 75 mg/m2 on day 2. These treatments should be repeated every 21 days and continued for as many as 6 cycles (with appropriate dose adjustment for cisplatin-related nonhematologic toxicities) as long as neuropathy does not exceed grade 1. With grade 2 neuropathy, the paclitaxel should be discontinued. If an IP port has not been placed, my policy would be to administer IV carboplatin plus paclitaxel, and then proceed as indicated in the clinical vignette. Lack of clinical trials (level 1) evidence in such a recommendation is troubling; even more troubling, however, is the inability to formulate an adequate treatment plan for a patient who does not fit into a clinical-trial algorithm. Further thoughts on additional situations in which IP therapy should be considered are explored in the next section.

When Should We Consider IP Drug Administration?

The weight of the evidence supports a favorable effect on outcome with the administration of IP cisplatin. In phase III trials, a favorable outcome has been demonstrated in stage III ovarian cancer that has been cytoreduced to < 2 cm residuum in one trial and < 1 cm in two trials. All utilized IP cisplatin at 100 mg/m2 per cycle to be given over 6 cycles, but completion rates and platinum dose intensity were considerably less in these three trials-nevertheless, they achieved superior results! Toxicity considerations and trials by the IV route suggest that even a reduced dose of IP cisplatin at 50 mg/m2 achieves results comparable to higher doses given to similar total cumulative amounts.[45] Currently, an IP cisplatin dose of 75 mg/m2 could be used in patients who meet the criteria of the previously described debulking characteristics and in whom an IP catheter has been inserted, with the expectation of achieving results similar to those of GOG172.

Suboptimally debulked patients and patients in whom an IP port has not been placed should also be considered for IP therapy after documentation of complete regression by imaging and CA125 after 4 cycles of IV carboplatin plus paclitaxel. A reassessment no later than cycle 4 (and before 6 cycles are administered) may be vastly more satisfactory for consolidating with IP cisplatin, for the following reasons: 1) any advantage from extending IV doublet treatment or even crossing over to other IV drugs beyond 4 cycles has not been proven; 2) all evidence supporting IP treatment suggests it is effective before occurrence of platinum resistance; and 3) if residual peritoneal disease is documented at such reassessment, at least four more cycles of IP therapy could be desirable and for toxicity reasons, very challenging to administer after 6 cycles of IV therapy (and even worse if the patient has received 8 IV cycles, as given by some without any evidence favoring such lengthy treatments).

Should IP therapy be contemplated as consolidation now that IV bevacizumab maintenance therapy shows an improved PFS after first-line therapy? There is a good preclinical rationale for antiangiogenic agents to potentiate IP drug administration, both for cisplatin and for topotecan,[46] but there are not enough tolerance studies at present for the safety of this treatment, and one must await the results of GOG252.[45]

Another scenario in which IP therapy might be considered is in the recurrence setting, when the recurrence is peritoneal and is either low-volume disease or can be easily debulked. The initial case series by King et al[47] and several phase II studies point to considering such an intervention under these circumstances. Below we add clinical vignette numbers 2 and 3. Clinical vignette number 2 also shows how drugs that are not effective by the IV route may have activity against peritoneal disease.

Clinical vignette 2-A 68-year-old woman originally diagnosed in 1979 was referred in 1981 to Dr. Stephen Howell, to enroll in an IP cytarabine (ARA-C, Cytosar) protocol.[47] She had just suffered here second recurrence of cytology-positive ascites after a few months in remission following cisplatin-containing regimens. She received 6 cycles of cytarabine, and remained in documented clinical complete remission prior to her death 9 years later from pneumonia complicating Alzheimer’s disease during her last 2 years.

Clinical vignette 3-This woman, mother of two daughters, is now 70 years old. Her initial diagnosis was made in 1992, as stage IIIB high-grade papillary serous cancer following optimal cytoreductive surgery upon demonstration of an elevated CA125 (obtained by her gynecologist at the patient’s request). One year following completion of 6 cycles of caboplatin + cyclophosphamide, she experienced a symptomatic pelvic recurrence with presence of a cul-de-sac mass and pelvic ascites. She underwent surgical resection and insertion of an IP catheter, and entered a phase I study of IP platinums combined with floxuridine.[27] She received 6 cycles of IP cisplatin plus floxuridine and remains well, with no recurrence 16 years after cisplatin plus FUDR.

Finally, patients with synchronous ovarian and uterine primaries and those with uterine papillary serous cancer may also potentially benefit from IP approaches, as illustrated by clinical vignette 4.

Clinical vignette 4-This 49-year-old woman was diagnosed with synchronous ovarian and endometrial papillary serous primaries. She was optimally debulked and staged by transabdominal hysterectomy, bilateral salpingo-oophorectomy, and omentectomy and lymph node dissection on March 25, 2005. Her diagnosis had been preceded by finding an abnormal Pap test result during a routine exam in February 2005. In consultation, author FM reinforced concurrence with the planned 6 cycles of carboplatin plus paclitaxel systemic treatment, but given the patient’s history and the pathology, he discussed at some length the desirability of IP consolidation. Her pelvic cytology from the surgery was positive, making the ovarian cancer stage IIC, while the endometrial cancer was stage IA and grade I with < 5% focal high-grade component. IP therapy was recommended and subsequently, with a negative second-look surgery, the patient was given 2 cycles of IP cisplatin as 60 mg/m2 as consolidation therapy. She remains disease-free at last follow-up in November 2010.

Conclusion

The place of IP chemotherapy in the treatment of ovarian cancer continues to be broadly discussed, but few appreciate the many nuances of this wide-ranging debate. For precisely this reason, it is important to emphasize the key role and rationale for IP administration of cisplatin, which may be lost amongst the many concerns about IP drug delivery and its toxicity. Our major premise within this debate is that this successful treatment strategy should be kept simple, to ensure safety while retaining the positive signal coming from the trials reviewed (ie, based on IP cisplatin at doses ranging from 60–75 mg/m2, depending on clinical circumstances and prior exposure to platinum drugs). Of interest, a meta-analysis of treatments for ovarian cancer points to IP therapy as leading to yet another incremental advance.[48,49]

Ongoing clinical trials by the GOG and others will undoubtedly clarify these issues that have been raised, and they hold promise for enhancement in efficacy (eg, by adding bevacizumab) or reductions in toxicity (eg, based on findings with IP carboplatin). The results of these trials are eagerly awaited. In the meantime, IP therapy should be considered on an individual basis in patients who do not qualify for clinical trials. A strategy utilizing IP cisplatin administration is already applicable to many patients, based on the weight of the evidence to date.

Financial Disclosure:The authors have no significant interest or other relationship with the manufacturers of any products or providers of any service mentioned in this article.

Related Videos
Brian Slomovitz, MD, MS, FACOG discusses the use of new antibody drug conjugates for treating patients with various gynecologic cancers.
Developing novel regimens may continue to improve survival outcomes of patients with advanced cervical cancer following the FDA approval of pembrolizumab and chemoradiation, says Jyoti S. Mayadev, MD.
Treatment with pembrolizumab plus chemoradiation appears to be well tolerated with no detriment to quality of life among those with advanced cervical cancer.
Jyoti S. Mayadev, MD, says that pembrolizumab in combination with chemoradiation will be seamlessly incorporated into her institution’s treatment of those with FIGO 2014 stage III to IVA cervical cancer following the regimen’s FDA approval.
Domenica Lorusso, MD, PhD, says that paying attention to the quality of chemoradiotherapy is imperative to feeling confident about the potential addition of pembrolizumab for locally advanced cervical cancer.
Guidelines from the Society of Gynecologic Oncology may help with managing the ongoing chemotherapy shortage in the treatment of patients with gynecologic cancers, according to Brian Slomovitz, MD, MS, FACOG.
Interim data reveal favorable responses in patients with low-grade serous ovarian cancer treated with avutometinib plus defactinib, according to Susana N. Banerjee, MD.
Brian Slomovitz, MD, MS, FACOG, notes that sometimes there is a need to substitute cisplatin for carboplatin, and vice versa, to best manage gynecologic cancers during the chemotherapy shortage.
Findings from the phase 3 MIRASOL trial support mirvetuximab soravtansine as a standard treatment option for platinum-resistant ovarian cancer, according to Ritu Salani, MD.
Trastuzumab deruxtecan appears to elicit ‘impressive’ responses among patients with HER2-positive gynecologic cancers regardless of immunohistochemistry in the phase 2 DESTINY-PanTumor02 trial.
Related Content