Download Intraoperative Hyperthermic Intraperitoneal

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Patients With Advanced Ovarian Cancer
Review Article [1] | September 15, 2015 | Oncology Journal [2], Gynecologic Cancers [3], Ovarian
Cancer [4]
By Anton Oseledchyk, MD [5] and Oliver Zivanovic, MD, PhD [6]
Here we discuss the advantages and pitfalls of HIPEC in advanced ovarian cancer, as well as current
data and ongoing prospective trials.
Introduction
Ovarian cancer causes more deaths than any other gynecologic malignancy, accounting for 14,180
deaths in the United States annually, and with a prevalence of 21,290 new cases estimated for
2015.[1] Cure rates vary between 20% and 30%, with most patients experiencing recurrence of their
disease within the first 3 years. The reason for the poor prognosis is that the majority of patients
(60% to 70%) present at an advanced stage (International Federation of Gynecology and Obstetrics
[FIGO] stage III or IV).[2,3] Beyond stage, histology, and grade, a high prognostic value is attributed
to the amount of postoperative residual tumor.[4-7] Optimal cytoreduction, defined as residual
tumor < 1 cm after surgical debulking, is associated with considerable improvement in overall
survival (OS) and progression-free survival (PFS). More recent publications have demonstrated that a
complete resection of tumor with only microscopic tumor residuals will improve the prognosis to a
median OS of 64 months.[8] The goal of current cytoreductive surgery should therefore be to
attempt a complete resection of all visible tumor, and it is preferable that such surgery be performed
at centers with high levels of specialization and expertise.[4,9] Even so, and despite improvements
in surgical technique, most patients will die from ovarian cancer regardless of their increased
disease-free interval, making the improvement of chemotherapy regimens imperative.
Cisplatin’s remarkable activity in ovarian cancer was discovered in the early 1980s, followed by the
introduction of the less toxic but equally active second-generation platinum agent carboplatin in the
early 1990s. Both agents were predominantly used in combination regimens along with
cyclophosphamide or an anthracycline—until the introduction of the taxanes in the late 1990s led to
today’s gold standard of chemotherapy that combines carboplatin and paclitaxel in a 3-week
cycle.[10]
Biologically, ovarian cancer behaves in a unique way. After undergoing an epithelial-mesenchymal
transition, cancer cells simply detach from the main tumor bulk and are carried by the physiologic
peritoneal circulation throughout the peritoneal cavity, either as single cells or as multicellular
spheroids—something one could call “passive” metastasis.[11] The cells do not undergo several
steps of intravasation and extravasation to form metastases as in hematologically spreading tumor
types. In fact, ovarian cancer cells seem to selectively invade the mesothelium of the peritoneal
surface, forming micrometastases that are adequately supplied solely by means of diffusion until
they reach a size of 1 mm2.[12,13] Consequently, because of the lack of vascularization, it is likely
that neither micrometastases nor free-floating cancer cells can be addressed adequately by either
surgery or systemic chemotherapy. Intraperitoneal (IP) treatment modalities attempt to close this
therapeutic gap.
IP Chemotherapy: Rationale
In 1978, Dedrick et al hypothesized that because the peritoneal surface forms a barrier between the
peritoneal compartment and the blood compartment, it would be possible for significantly higher
concentrations of chemotherapeutic agents to be delivered to the peritoneal cavity, with only limited
systemic toxicity[14]; this hypothesis set the stage for the introduction of modern IP treatments. Any
drug that shows a high response rate as systemic therapy for ovarian cancer comes under
consideration for IP administration. However, there are important pharmacokinetic differences
between the different drugs used for IP therapy; these play a role in drug selection and therefore
Page 1 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
must be mentioned. There is a correlation between molecular size and the ratio of the drug level in
the peritoneal cavity to the drug level in plasma. For example, peak paclitaxel concentrations in the
peritoneal cavity exceed plasma concentrations by 1,000-fold and persist in the peritoneal cavity for
over 24 hours due to the large size of the paclitaxel molecule compared with cisplatin; the latter
shows only a 12-fold higher concentration in the peritoneal compartment compared with the
concentration in serum.[15] The significantly higher IP drug concentration might reduce the effect of
chemotherapy resistance by simply achieving higher intracellular concentrations and, in the case of
cisplatin, by overpowering the mechanisms of decreased drug influx that result from loss of copper
transporter 1, increased absorption of cisplatin in the cytoplasm by glutathione and metallothionein,
increased drug efflux by ATP7A/ATP7B and glutathione S-conjugate export GS-X pumps, improved
DNA repair, and insufficient DNA binding.[16]
Most studies of IP chemotherapy in ovarian cancer have involved cisplatin, because of positive
results with this agent in murine models and more experience with it as a systemic therapy.
However, recent prospective studies were able to demonstrate the feasibility of IP carboplatin
application,[17,18] which is an appealing alternative to cisplatin because of its lesser side effects,
especially less polyneuropathy and nephrotoxicity.
IP paclitaxel infusion showed dose-limiting toxicity (abdominal pain) at 175 mg/m2 in a phase I
trial.[19] Subsequent studies of a weekly IP regimen at 60 mg/m2 showed acceptable toxicity,[20,21]
with the result that a phase II trial combining IP cisplatin and IP paclitaxel was performed,[22]
demonstrating both feasibility and a high median OS. Because of the positive results, the
combination of IP cisplatin and IP paclitaxel was tested as the study arm in the phase III Gynecologic
Oncology Group (GOG) 172 trial described further on in detail. It is unclear whether the increased
adverse effects of paclitaxel are Cremophor EL–related, since the hydrophobic paclitaxel requires it
as a vehicle. It has been shown that Cremophor EL can have biologic effects such as severe
anaphylactic hypersensitivity reactions, abnormal lipoprotein patterns, and peripheral
neuropathy.[23] The use of water-soluble taxanes, such as docetaxel or the albumin-bound
nab-paclitaxel, might prevent these adverse effects in the future.
IP Chemotherapy: Clinical Significance
GOG 104, the first phase III trial to evaluate the role of IP treatment, showed a significant
improvement in OS: 49 months in the IP cisplatin treatment group vs 41 months in patients who
received intravenous (IV) treatment only.[24] Because the participants in this study were recruited in
the pre-paclitaxel era, survival rates were rather low compared with the rates in current adjuvant
chemotherapy studies. In the subsequent GOG 114 trial, patients were randomly assigned to receive
either 6 cycles of cisplatin and paclitaxel IV therapy or 6 cycles of the combination of paclitaxel IV
plus cisplatin IP after 2 previous cycles of carboplatin (area under the curve [AUC], 9),[25] with
significant improvement in median PFS (28 months vs 22 months) and close to significant
improvement in median OS (63 months vs 52 months) favoring the IP regimen. The most recently
published phase III trial, GOG 172, randomly assigned patients—after primary debulking of stage III
ovarian, fallopian tube, or peritoneal cancer with postoperative residual tumor < 1 cm—to receive
either IV paclitaxel, 135 mg/m2 over 24 hours, on day 1 and IV cisplatin, 75 mg/m2, on day 2, or IV
paclitaxel, 135 mg/m2 over 24 hours, on day 1 and IP cisplatin, 100 mg/m2 over 24 hours, on day 2,
followed by IP paclitaxel, 60 mg/m2, on day 8 of a 3-week cycle. The investigators showed a
significantly increased PFS and OS of 23.8 months vs 18.3 months and 65.5 months vs 49.7 months,
respectively, favoring the IP regimen. Based on these results, the National Cancer Institute issued an
alert encouraging the incorporation of IP therapy into the care of women with advanced ovarian
cancer in the United States.[26]
However, the strong effect of the IP regimens in GOG 114 and GOG 172 may have been due to the
higher dosing in the IP treatment arms: 2 additional IV carboplatin cycles as well as a higher IP
cisplatin dose in GOG 114; and an additional dose of paclitaxel on day 8 of every cycle in GOG 172,
resembling a dose-dense IV regimen. Additionally, 44% of patients in the IP treatment arm of GOG
172 received IV carboplatin and paclitaxel after discontinuing the IP protocol. This IV regimen may
have been less toxic and more effective than the IV regimen used in the control arm (paclitaxel on
day 1 and cisplatin day 2), which itself does not resemble today’s standard-of-care treatment.
Because of these issues and toxicity concerns, as well as an only marginally significant OS benefit in
the GOG 172 trial, an opposing statement was published in the Journal of Clinical Oncology in
October 2006 advising against IP therapy outside of clinical trials.[27]
Recently, Tewari et al[28] presented an updated long-term survival analysis of GOG 114 and GOG
Page 2 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
172, with a median follow-up of 10.7 years. Median OS with IP therapy was 61.8 months, compared
with 51.4 months in the IV group, with a reduction in the risk of death of 23%. Additionally, the
number of IP cycles revealed itself as an independent prognostic factor: completing all 6 cycles had
a statistically significant association with improved OS, whereas completion of fewer than 6 cycles of
IP therapy did not. IP therapy also improved the survival of patients with gross residual disease (≤ 1
cm); for this reason, patients with suboptimal debulking have been included in the ongoing GOG 252
trial. GOG 252 compares the following three treatment groups: conventional dose-dense IV
chemotherapy, dose-dense IV chemotherapy with IP (instead of IV) carboplatin delivery, and a
modified GOG 172 IP protocol. In all treatment arms, patients have received 22 cycles of
every-3-weeks bevacizumab, 15 mg/kg. The first results of GOG 252 are anticipated either this year
or next.
Challenges Regarding Establishment of IP Chemotherapy as Standard of
Care
The major disadvantages of IP chemotherapy are the increased toxicity and the highly complex
management of patients and their side effects. In GOG 172, 58% of patients discontinued therapy
due to increased hematologic and gastrointestinal toxicity; inadequate hydration or inadequate
antiemetic therapy; or port complications, including obstruction, leakage, and infection.[29,30]
Additionally, Havrilesky et al, using a Markov state transition model, showed that IP treatment was
not cost-effective compared with IV therapy, mainly because of the need for inpatient treatment for
the 24-hour delivery of paclitaxel.[31] Both the 24-hour paclitaxel infusion in the GOG 172 protocol
and the increased adverse effects of IP chemotherapy have been addressed by a modified GOG 172
outpatient regimen that was developed at Memorial Sloan Kettering Cancer Center (MSKCC) in
recent years: IV paclitaxel, 135 mg/m2, infused on day 1 within 3 hours, followed by reduced IP
cisplatin (75 mg/m2) and IP paclitaxel (60 mg/m2) on day 8, along with implementation of new potent
antiemetic drugs (eg, aprepitant). This approach has been validated in a single-arm phase II study
that showed low discontinuation rates[32]: 30 patients (73%) received all 6 cycles of IP
chemotherapy and 35 patients (85%) received at least 4 cycles. The greatly reduced toxicity of this
regimen is reassuring, given that it is one of the treatment arms of the ongoing GOG 252 trial.
Independent of whether the outpatient protocol will have a comparable efficacy to that of the GOG
172 regimen, IP treatment will still be challenging for healthcare providers due to higher
complication rates, the need for additional homecare to ensure adequate IV hydration, longer
treatment times, and intensified nurse involvement. These factors remain the major limitations
impeding the establishment of IP chemotherapy as the standard of care.
HIPEC: Rationale
In recent years, a new form of IP therapy has emerged for patients with ovarian carcinoma:
intraoperative hyperthermic IP chemotherapy (HIPEC). Many investigators are now evaluating and
conducting critical discussions of the role and the rationale for this delivery technique, which
requires intraoperative perfusion machines, elaborate logistics, and a high degree of organizational
effort. It is still unknown whether HIPEC is associated with an improved survival that would justify the
effort involved, but there are several potential advantages that make it a promising therapeutic
option as part of a multimodality treatment:
• A high volume of chemotherapy can be delivered, and a homogenous distribution can be achieved.
This is often not practical in conventional IP therapy, because of abdominal distension and pain, but
it is feasible in HIPEC, since the patient is under anesthesia.
• There is no interval between cytoreduction and chemotherapy. The cytotoxic therapy is applied at
the time of minimal disease manifestation, and there are no adhesions that might alter the
distribution of the drug.
• Hyperthermia has a pharmacokinetic benefit. Several studies have convincingly shown that
hyperthermia can increase both the tumor penetration of cisplatin[33] as well as the DNA
crosslinking.[34]
• High concentrations of chemotherapy can be achieved in the intraperitoneal compartment with low
systemic exposure—in a single intraoperative treatment.
HIPEC: Clinical Significance
HIPEC is already an established treatment alternative in three tumor types, based on some
Page 3 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
prospective evidence and a large number of retrospective studies: peritoneal carcinomatosis of
colorectal cancer, appendiceal cancer (pseudomyxoma peritonei), and malignant peritoneal
mesothelioma. Two randomized trials from France and the Netherlands[35,36] have shown a
significant improvement in OS with HIPEC in peritoneal dissemination of colorectal cancer—albeit
with a major limitation: the control arms in these studies did not resemble the current gold standard
of oxaliplatin-based[37,38] or irinotecan-based therapy[39] (FOLFOX [folinic acid, fluorouracil, and
oxaliplatin] or FOLFIRI [folinic acid, fluorouracil, and irinotecan], respectively). Nevertheless,
compared with fluorouracil (5-FU) and folinic acid (leucovorin), HIPEC improved median OS from 10
months to 29 months, especially favoring patients with complete cytoreduction (in whom OS
improved from 28 months to 60 months).[40] In pseudomyxoma peritonei, there is evidence of a
substantial benefit from HIPEC[41,42]; however, there have been no randomized trials of HIPEC in
this entity, and it is unclear whether the improved outcome is attributable to the successful
cytoreduction at the highly specialized centers at which studies have been performed or whether it is
actually due to the addition of the IP chemotherapy. In malignant peritoneal mesothelioma, HIPEC
has been accepted as the first-line treatment, despite the lack of randomized trials. Large
retrospective studies have shown improved OS compared with historical controls.[43,44] In all the
above studies in various malignancies, a compelling benefit was shown for patients with a complete
cytoreduction. Attention must now be focused on the issue of patient selection for this very involved
procedure, through the use of objectified tumor volume scores, such as the peritoneal cancer index
(PCI).[45]
In recent years, HIPEC has been studied in ovarian cancer; however, due to the lack of randomized
trials there are no substantive efficacy data. The largest retrospective study in persistent and
recurrent ovarian cancer, by Bakrin et al, described survival and morbidity in 246 patients over a
period of 17 years,[46] and showed both acceptable morbidity (12%) and a median OS of 48.9
months. Still, this study has substantial limitations that cannot be ignored: the use of different
postoperative treatment regimens over the 17-year time period (1991 to 2008), high complete
resection rates of 92% (which are usually not achieved even by leading institutions), and the
inclusion of platinum-resistant and platinum-sensitive disease.[47] Despite its weaknesses, the study
proves the feasibility of the procedure and encourages prospective randomized trials. At the same
time, however, this study demonstrates an unfortunate development in the treatment of ovarian
cancer patients: more than 500 patients in this and other smaller retrospective evaluations have
been treated with HIPEC outside a prospective study protocol, without control arms and
consequently without proof of efficacy, indicating a wide application of a yet immature and not
validated treatment modality.
TO PUT THAT INTO CONTEXT
David L. Bartlett, MD
University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
The concept of intraperitoneal (IP) chemotherapy for peritoneal metastases makes intuitive sense: it
maximizes the concentration of chemotherapy delivered to the tumor cells while minimizing
systemic toxicity. Randomized trials have demonstrated a significant and meaningful benefit to IP
dwell therapy, but this approach also has significant limitations: technical complexity, pain for the
patient, increased toxicity, and limited distribution of the drug due to adhesions after surgery.
WHY HIPEC?
In an attempt to address these limitations, gastrointestinal (GI) surgical oncologists began to use
hyperthermic IP chemotherapy (HIPEC) in the early 1990s. This approach has theoretical advantages
over IP dwell therapy: it is delivered under general anesthesia at the time of cytoreduction; it utilizes
hyperthermia, which has direct cytotoxic effects and which potentiates chemotherapy; and it
Page 4 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
virtually assures complete distribution of drug throughout the abdominal cavity. What makes sense
for GI tumors makes even more sense for ovarian cancer, since the latter is more chemosensitive.
This means that a brief exposure to cisplatin and hyperthermia can have dramatic effects.
WHY IS HIPEC STILL CONTROVERSIAL?
After almost 25 years of data, however, the utility of HIPEC is not uniformly accepted, techniques
vary, drugs and dosages differ, and we have no idea which aspect of the treatment (cytoreduction,
hyperthermia, or chemotherapy) makes it effective.
WHAT DOES THE FUTURE HOLD?
It is exciting to see the results of one randomized trial demonstrating a greater than doubling of
median survival in patients with recurrent ovarian cancer. It is even more encouraging to see that
seven randomized trials examining HIPEC are ongoing in ovarian cancer. The results of these
ongoing trials will help answer important questions regarding the efficacy of HIPEC and the most
appropriate indications for its use. The implications for clinical practice will likely be significant.
For this reason, many groups are advising against the use of HIPEC in ovarian cancer patients
outside of clinical trials.[48] Two main criticisms are dominating the discussion: OS data are missing,
and complication rates are higher than with conventional surgical treatment. Although there is no
question that the lack of OS data is a problem, there are convincing studies that refute safety and
feasibility concerns regarding this treatment modality. A number of phase I trials were conducted in
both primary[49,50] and recurrent disease.[50,51] Severe complications ranged between 15% and
25% and morbidity between 0% and 4.2%. In one of these trials, even heavily pretreated patients
with recurrent disease who had already undergone extensive surgery and adjuvant chemotherapy
tolerated secondary cytoreductive surgery and HIPEC with low complication rates, no deaths, no
delay in the initiation of standard postoperative carboplatin and gemcitabine chemotherapy, and no
increased chemotherapy-associated adverse effects.[51] This dose-finding study showed that high IP
cisplatin concentrations (100 mg/m2) are feasible in pretreated patients.
Much as with postoperative IP chemotherapy, paclitaxel has only recently been investigated in the
setting of HIPEC. With the peritoneal uptake of paclitaxel significantly slower than that of
cisplatin,[52] an increased antitumor effect has been shown in vitro.[53] In a single-arm phase I
study, the delivery of paclitaxel, 175 mg/m2, in the setting of HIPEC showed acceptable morbidity
(38%; minor and major complications), with no postoperative deaths.[54] Ansaloni et al conducted a
multi-regimen phase II trial, including 11 patients who received cisplatin and paclitaxel in the setting
of HIPEC; however, the authors did not provide complication rates for this specific regimen.[55] A
pharmacokinetic evaluation of concurrent cisplatin and paclitaxel in HIPEC indicated that the serum
concentration of paclitaxel would be below that associated with the regimen’s dose-limiting toxicity
yet would constitute an effective IP concentration.[56]
HIPEC: Randomized Trials
A single-institution randomized phase III trial by Spiliotis et al was recently published that compared
conventional cytoreduction in a first recurrence with cytoreduction and HIPEC (with cisplatin, 100
mg/m2, and paclitaxel, 175 mg/m2, in platinum-sensitive disease; and doxorubicin, 35 mg/m2, and
paclitaxel, 175 mg/m2, or mitomycin, 15 mg/m2, in platinum-resistant disease).[57] The authors
demonstrated a significant improvement in the mean OS: 29.7 months in the HIPEC arm vs 13.4
months in the surgery-only arm (P = .006). Interestingly, patients with platinum-resistant disease
showed a stronger improvement in median OS, while still having a significantly impaired OS
compared with the OS seen in patients with platinum-sensitive disease. As anticipated, the highest
OS was observed in patients with complete cytoreduction who received HIPEC; in addition, the
preoperative tumor burden as reflected in the PCI score was described as an independent prognostic
factor, with PCI score > 15 associated with a significantly impaired survival. Unfortunately, there are
several weaknesses in the presentation of the data that decrease the validity of this first randomized
HIPEC trial: There is no information on PFS, the authors do not provide median follow-up data, and
the Kaplan-Meier survival curve shows a high number of censored cases. There is no information
regarding the postoperative first-line treatment, nor are the complication rates addressed. Also,
different regimens were used in patients with platinum-sensitive and platinum-resistant disease.
Currently, there are six open randomized trials (Table) recruiting patients with both primary and
recurrent disease, as well as women who have already received 3 cycles of neoadjuvant
chemotherapy. The CARCINOHIPEC study has completed recruitment, and the first results are
anticipated soon. These studies will certainly provide more useful information about this treatment
Page 5 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
modality.
HIPEC: Future Outlook
As unclear as the patient outcome benefit may be, HIPEC is highly important as a platform for
studying the effect of IP chemotherapy and hyperthermia on cancer cells in vivo. It offers the unique
possibility of evaluating macroscopic lesions within the peritoneal cavity during chemoperfusion and
then harvesting them after the procedure. The application of gene expression and proteomics to
biopsy specimens could shed new light on the escape mechanisms of cancer cells and mechanisms
of drug resistance. Pre- and post-perfusion biopsies could facilitate human in vivo studies of either
new agents for IP therapy or methods for the improvement of chemotherapy uptake, thereby
accelerating the process by which new therapeutics become available for clinical use. Two of the
most promising emerging approaches are nanoparticle delivery systems and immunotherapy.
Nanoparticle delivery of paclitaxel and cisplatin has been the subject of recent clinical studies. The
chief rationale for nanoparticle-facilitated IP chemotherapy is reduced clearance of the
nanoparticle-bound drug from the peritoneal cavity compared with the clearance of a conventional
drug in solution, and because of that, a higher peritoneum-to-plasma ratio as well as reduced
systemic effects and longer tumor exposure. Especially for the small-molecule and water-soluble
drugs cisplatin and carboplatin, reduced peritoneal clearance would result in a higher diffusion
gradient and thus enhanced tumor penetration. In a murine model, paclitaxel has been successfully
incorporated into nanoparticles,[58] in order to achieve higher tumor selectivity after IP injection and
longer retention times. It was combined with yttrium-90 as part of a multimodal treatment.[58]
Instead of incorporating paclitaxel into a nanoparticle, a new synthesis technique called precipitation
with compressed antisolvent (PCA) allows the production of 800 nm–sized paclitaxel crystals that do
not require use of the toxic solvent Cremophor EL; this technique has shown promising results after
IP injection in a murine model.[59] A phase I trial was completed and the results are anticipated.
Clinical trials of nanoparticle delivery systems for cytotoxic drugs have until now involved
predominantly IV injection, with two agents that use this mode of delivery approved by the US Food
and Drug Administration: nanoparticle albumin-bound paclitaxel and pegylated liposomal
doxorubicin.[60,61] Other nanoparticles incorporating paclitaxel[62] or platinum polymers[63] for IV
injection are under clinical investigation. A phase III noninferiority trial of a novel water-soluble
formulation of paclitaxel and XR-17 (paclical) has been completed and was presented at the 2015
American Society of Clinical Oncology Annual Meeting.[64] The trial showed comparable rates of
adverse effects and PFS; however, contrary to expectations, Cremophor EL–related adverse effects,
such as neuropathy and allergic reactions, were not significantly reduced. Paclical has been
approved for use in the Russian Federation, but approvals in the United States and the European
Union are still pending.
In addition to their use for the delivery of cytotoxic agents, nanoparticles can be utilized to
encapsulate other substances that are unstable in serum or ascites, releasing agents such as small
interfering RNAs (siRNAs) into the cell cytoplasm of cancer cells.[65] siRNAs can silence
overexpressed genes and reduce tumor growth and invasion, with low toxicity. The use of
nanoparticles to deliver siRNAs has recently been shown to be applicable to humans.[66] Major
targets for siRNAs in ovarian cancer are the genes encoding folate receptor, follicle-stimulating
hormone receptor, luteinizing hormone–releasing hormone receptor, mucin 1, and epidermal growth
factor receptor.[67]
Another new approach is IP immunotherapy, which involves the delivery of programmed T cells.
Since Zhang et al showed that tumor infiltration with CD8-positive lymphocytes is associated with
improved 5-year survival in epithelial ovarian cancer,[68] several attempts have been made to
modify the T-cell response.[69] This can be achieved either by harvesting cells, reprogramming them
in vitro, and then reintroducing the cells via autologous injection,[70,71] or by stimulating T-cell
activation in vivo.[72] There are several ongoing phase I trials, but the efficacy as well as the
relevance of IP applications are unclear.
Conclusion
The rationale for HIPEC as part of a multimodal treatment in patients with advanced ovarian cancer
is strong. Given that hyperthermia enhances tumor penetration and the cytotoxic effects of
chemotherapy, recent improvements in drug distribution and in patient monitoring in the operating
room, as well as encouraging results in nonrandomized trials, justify further investigation of HIPEC in
randomized clinical trials. The possibility of easily acquiring pre- and post-treatment biopsies in the
Page 6 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
course of the procedure also makes HIPEC an excellent setting for human in vivo studies of
antitumor effects and pharmacodynamics.
Financial Disclosure: The authors have no significant financial interest in or other relationship with
the manufacturer of any product or provider of any service mentioned in this article.
Table. Ongoing Prospective Clinical Trials
Evaluating the Role of HIPE...
References:
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5-29.
2. Kaatsch P, Spix C, Hentschel S, et al. Krebs in Deutschland 2009/2010 - 9. Ausgabe, 2013.
http://www.rki.de/Krebs/DE/Content/Publikationen/Krebs_in_Deutschland/kid_2013/krebs_in_deutschl
and_2013.pdf?__blob=publicationFile. Accessed August 19, 2015.
3. National Cancer Institute. Howlader N, Noone A, Krapcho M, et al, editors. SEER cancer statistics
review, 1975-2012. http://seer.cancer.gov/CSR/1975_2012. Accessed August 26, 2015.
4. Bristow RE, Tomacruz RS, Armstrong DK, et al. Survival effect of maximal cytoreductive surgery
for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol.
2002;20:1248-59.
5. Chi DS, Eisenhauer EL, Lang J, et al. What is the optimal goal of primary cytoreductive surgery for
bulky stage IIIC epithelial ovarian carcinoma (EOC)? Gynecol Oncol. 2006;103:559-64.
6. Winter WE 3rd, Maxwell GL, Tian C, et al. Prognostic factors for stage III epithelial ovarian cancer:
a Gynecologic Oncology Group study. J Clin Oncol. 2007;25:3621-7.
7. Eisenkop S, Friedman R, Wang H. Complete cytoreductive surgery is feasible and maximizes
survival in patients with advanced epithelial ovarian cancer: a prospective study. Gynecol Oncol.
1998;69:103-8.
8. Winter WE 3rd, Maxwell GL, Tian C, et al. Tumor residual after surgical cytoreduction in prediction
of clinical outcome in stage IV epithelial ovarian cancer: a Gynecologic Oncology Group study. J Clin
Oncol. 2008;26:83-9.
9. Giede KC, Kieser K, Dodge J, Rosen B. Who should operate on patients with ovarian cancer? An
evidence-based review. Gynecol Oncol. 2005;99:447-61.
10. Ozols RF, Bundy BN, Greer BE, et al. Phase III trial of carboplatin and paclitaxel compared with
cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic
Oncology Group study. J Clin Oncol. 2003;21:3194-200.
11. Lengyel E. Ovarian cancer development and metastasis. Am J Pathol. 2010;177:1053-64.
12. Bamberger ES, Perrett CW. Angiogenesis in epithelian ovarian cancer. Mol Pathol.
Page 7 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
2002;55:348-59.
13. Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst.
1990;82:4-6.
14. Dedrick RL. Theoretical and experimental bases of intraperitoneal chemotherapy. Semin Oncol.
1985;12:1-6.
15. Fujiwara K, Armstrong D, Morgan M, Markman M. Principles and practice of intraperitoneal
chemotherapy for ovarian cancer. Int J Gynecol Cancer. 2007;17:1-20.
16. Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer.
2007;7:573-84.
17. Speyer JL, Sorich J. Intraperitoneal carboplatin: rationale and experience. Semin Oncol.
1992;19:107-13.
18. Gould N, Sill MW, Mannel RS, et al. A phase I study with an expanded cohort to assess the
feasibility of intravenous paclitaxel, intraperitoneal carboplatin and intraperitoneal paclitaxel in
patients with untreated ovarian, fallopian tube or primary peritoneal carcinoma: A Gynecologic
Oncology Group study. Gynecol Oncol. 2012;125:54-8.
19. Markman M, Rowinsky E, Hakes T, et al. Phase I trial of intraperitoneal Taxol: a Gynecoloic
Oncology Group study. J Clin Oncol. 1992;10:1485-91.
20. Francis P, Rowinsky E, Schneider J, et al. Phase I feasibility and pharmacologic study of weekly
intraperitoneal paclitaxel: a Gynecologic Oncology Group pilot study. J Clin Oncol. 1995;13:2961-7.
21. Markman M, Brady MF, Spirtos NM, et al. Phase II trial of intraperitoneal paclitaxel in carcinoma
of the ovary, tube, and peritoneum: a Gynecologic Oncology Group study. J Clin Oncol.
1998;16:2620-4.
22. Rothenberg ML, Liu PY, Braly PS, et al. Combined intraperitoneal and intravenous chemotherapy
for women with optimally debulked ovarian cancer: results from an intergroup phase II trial. J Clin
Oncol. 2003;21:1313-9.
23. Gelderblom H, Verweij J, Nooter K, Sparreboom A. Cremophor EL: the drawbacks and advantages
of vehicle selection for drug formulation. Eur J Cancer. 2001;37:1590-8.
24. Alberts DS, Liu PY, Hannigan EV, et al. Intraperitoneal cisplatin plus intravenous
cyclophosphamide versus intravenous cisplatin plus intravenous cyclophosphamide for stage III
ovarian cancer. N Engl J Med. 1996;335:1950-5.
25. Markman M, Bundy BN, Alberts DS, et al. Phase III trial of standard-dose intravenous cisplatin
plus paclitaxel versus moderately high-dose carboplatin followed by intravenous paclitaxel and
intraperitoneal cisplatin in small-volume stage III ovarian carcinoma: an intergroup study of the
Gynecologic Oncology Group, Southwestern Oncology Group, and Eastern Cooperative Oncology
Group. J Clin Oncol. 2001;19:1001-7.
26. Clinical advisory: NCI issues clinical announcement for preferred method of treatment for
advanced ovarian cancer. US National Library of Medicine; 03 Jan 2006.
http://www.nlm.nih.gov/databases/alerts/ovarian_ip_chemo.html. Accessed April 28, 2015.
27. Gore M, du Bois A, Vergote I. Intraperitoneal chemotherapy in ovarian cancer remains
experimental. J Clin Oncol. 2006;24:4528-30.
28. Tewari D, Java JJ, Salani R, et al. Long-term survival advantage and prognostic factors associated
with intraperitoneal chemotherapy treatment in advanced ovarian cancer: a Gynecologic Oncology
Group study. J Clin Oncol. 23 Mar 2015. [Epub ahead of print]
Page 8 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
29. Armstrong DK, Bundy B, Wenzel L, et al. Intraperitoneal cisplatin and paclitaxel in ovarian
cancer. N Engl J Med. 2006;354:34-43.
30. Walker JL, Armstrong DK, Huang HQ, et al. Intraperitoneal catheter outcomes in a phase III trial
of intravenous versus intraperitoneal chemotherapy in optimal stage III ovarian and primary
peritoneal cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2006;100:27-32.
31. Havrilesky LJ, Secord AA, Darcy KM, et al. Cost effectiveness of intraperitoneal compared with
intravenous chemotherapy for women with optimally resected stage III ovarian cancer: a
Gynecologic Oncology Group study. J Clin Oncol. 2008;26:4144-50.
32. Konner JA, Grabon DM, Gerst SR, et al. Phase II study of intraperitoneal paclitaxel plus cisplatin
and intravenous paclitaxel plus bevacizumab as adjuvant treatment of optimal stage II/III epithelial
ovarian cancer. J Clin Oncol. 2011;29:4662-8.
33. van de Vaart PJM, van der Vange N, Zoetmulder FAN, et al. Intraperitoneal cisplatin with regional
hyperthermia in advanced ovarian cancer: pharmacokinetics and cisplatin-DNA adduct formation in
patients and ovarian cancer cell lines. Eur J Cancer. 1998;34:148-54.
34. Los G, van Vugt MJ, den Engelse L, Pinedo HM. Effects of temperature on the interaction of
cisplatin and carboplatin with cellular DNA. Biochem Pharmacol. 1993;46:1229-37.
35. Verwaal VJ, van Ruth S, de Bree E, et al. Randomized trial of cytoreduction and hyperthermic
intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with
peritoneal carcinomatosis of colorectal cancer. J Clin Oncol. 2003;21:3737-43.
36. Elias D, Delperro JR, Sideris L, et al. Treatment of peritoneal carcinomatosis from colorectal
cancer: impact of complete cytoreductive surgery and difficulties in conducting randomized trials.
Ann Surg Oncol. 2004;11:518-21.
37. Giacchetti S, Perpoint B, Zidani R, et al. Phase III multicenter randomized trial of oxaliplatin
added to chronomodulated fluorouracil-leucovorin as first-line treatment of metastatic colorectal
cancer. J Clin Oncol. 2000;18:136-47.
38. de Gramont A, Figer A, Seymour M, et al. Leucovorin and fluorouracil with or without oxaliplatin
as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000;18:2938-47.
39. Douillard JY, Cunningham D, Roth AD, et al. Irinotecan combined with fluorouracil compared with
fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised
trial. Lancet. 2000;355:1041-7.
40. Yan TD, Black D, Savady R, Sugarbaker PH. Systematic review on the efficacy of cytoreductive
surgery combined with perioperative intraperitoneal chemotherapy for peritoneal carcinomatosis
from colorectal carcinoma. J Clin Oncol. 2006;24:4011-9.
41. Bryant J, Clegg AJ, Sidhu MK, et al. Systematic review of the Sugarbaker procedure for
pseudomyxoma peritonei. Br J Surg. 2005;92:153-8.
42. Chua TC, Moran BJ, Sugarbaker PH, et al. Early- and long-term outcome data of patients with
pseudomyxoma peritonei from appendiceal origin treated by a strategy of cytoreductive surgery and
hyperthermic intraperitoneal chemotherapy. J Clin Oncol. 2012;30:2449-56.
43. Helm JH, Miura JT, Glenn JA, et al. Cytoreductive surgery and hyperthermic intraperitoneal
chemotherapy for malignant peritoneal mesothelioma: a systematic review and meta-analysis. Ann
Surg Oncol. 2015;22:1686-93.
44. Yan TD, Deraco M, Baratti D, et al. Cytoreductive surgery and hyperthermic intraperitoneal
chemotherapy for malignant peritoneal mesothelioma: multi-institutional experience. J Clin Oncol.
Page 9 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
2009;27:6237-42.
45. Esquivel J, Farinetti A, Sugarbaker PH. [Elective surgery in recurrent colon cancer with peritoneal
seeding: when to and when not to proceed]. G Chir. 1999;20:81-6.
46. Bakrin N, Cotte E, Golfier F, et al. Cytoreductive surgery and hyperthermic intraperitoneal
chemotherapy (HIPEC) for persistent and recurrent advanced ovarian carcinoma: a multicenter,
prospective study of 246 patients. Ann Surg Oncol. 2012;19:4052-8.
47. Herzog TJ. The role of heated intraperitoneal chemotherapy (HIPEC) in ovarian cancer: hope or
hoax? Ann Surg Oncol. 2012;19:3998-4000.
48. Harter P, Mahner S, Hilpert F, et al. Statement by the Kommission OVAR of the AGO Study Group
on the use of HIPEC (hyperthermic intraperitoneal chemotherapy) to treat primary and recurrent
ovarian cancer. Geburtshilfe Frauenheilkd. 2013;73:221-3.
49. Deraco M, Kusamura S, Virzi S, et al. Cytoreductive surgery and hyperthermic intraperitoneal
chemotherapy as upfront therapy for advanced epithelial ovarian cancer: multi-institutional phase II
trial. Gynecol Oncol. 2011;122:215-20.
50. Di Giorgio A, Naticchioni E, Biacchi D, et al. Cytoreductive surgery (peritonectomy procedures)
combined with hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of diffuse
peritoneal carcinomatosis from ovarian cancer. Cancer. 2008;113:315-25.
51. Zivanovic O, Abramian A, Kullmann M, et al. HIPEC ROC I: a phase I study of cisplatin
administered as hyperthermic intraoperative intraperitoneal chemoperfusion followed by
postoperative intravenous platinum-based chemotherapy in patients with platinum-sensitive
recurrent epithelial ovarian cancer. Int J Cancer. 2015;136:699-708.
52. Kamei T, Kitayama J, Yamaguchi H, et al. Spatial distribution of intraperitoneally administrated
paclitaxel nanoparticles solubilized with poly (2-methacryloxyethyl phosphorylcholine-co n-butyl
methacrylate) in peritoneal metastatic nodules. Cancer Sci. 2011;102:200-5.
53. Urano M, Kuroda M, Nishimura Y. For the clinical application of thermochemotherapy given at
mild temperatures. Int J Hyperthermia. 1999;15:79-107.
54. de Bree E, Rosing H, Filis D, et al. Cytoreductive surgery and intraoperative hyperthermic
intraperitoneal chemotherapy with paclitaxel: a clinical and pharmacokinetic study. Ann Surg Oncol.
2008;15:1183-92.
55. Ansaloni L, Agnoletti V, Amadori A, et al. Evaluation of extensive cytoreductive surgery and
hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with advanced epithelial ovarian
cancer. Int J Gynecol Cancer. 2012;22:778-85.
56. Ansaloni L, Coccolini F, Morosi L, et al. Pharmacokinetics of concomitant cisplatin and paclitaxel
administered by hyperthermic intraperitoneal chemotherapy to patients with peritoneal
carcinomatosis from epithelial ovarian cancer. Br J Cancer. 2015;112:306-12.
57. Spiliotis J, Halkia E, Lianos E, et al. Cytoreductive surgery and HIPEC in recurrent epithelial
ovarian cancer: a prospective randomized phase III study. Ann Surg Oncol. 2015;22:1570-5.
58. Werner ME, Karve S, Sukumar R, et al. Folate-targeted nanoparticle delivery of chemo- and
radiotherapeutics for the treatment of ovarian cancer peritoneal metastasis. Biomaterials.
2011;32:8548-54.
59. Roby KF, Niu F, Rajewski RA, et al. Syngeneic mouse model of epithelial ovarian cancer: effects
of nanoparticulate paclitaxel, Nanotax. Adv Exp Med Biol. 2008;622:169-81.
60. Wagner U, Marth C, Largillier R, et al. Final overall survival results of phase III GCIG CALYPSO trial
Page 10 of 11
Intraoperative Hyperthermic Intraperitoneal Chemotherapy in
Published on Cancer Network (http://www.cancernetwork.com)
of pegylated liposomal doxorubicin and carboplatin vs paclitaxel and carboplatin in
platinum-sensitive ovarian cancer patients. Br J Cancer. 2012;107:588-91.
61. Ferrandina G, Ludovisi M, Lorusso D, et al. Phase III trial of gemcitabine compared with
pegylated liposomal doxorubicin in progressive or recurrent ovarian cancer. J Clin Oncol.
2008;26:890-6.
62. Morgan MA, Darcy KM, Rose PG, et al. Paclitaxel poliglumex and carboplatin as first-line therapy
in ovarian, peritoneal or fallopian tube cancer: a phase I and feasibility trial of the Gynecologic
Oncology Group. Gynecol Oncol. 2008;110:329-35.
63. Rademaker-Lakhai JM, Terret C, Howell SB, et al. A phase I and pharmacological study of the
platinum polymer AP5280 given as an intravenous infusion once every 3 weeks in patients with solid
tumors. Clin Cancer Res. 2004;10:3386-95.
64. Vergote I, Brize A, Lisyanskaya AS, et al. Randomized phase III study comparing
paclical-carboplatin with paclitaxel-carboplatin in patients with recurrent platinum-sensitive epithelial
ovarian cancer. J Clin Oncol. 2015;33(suppl):abstr 5517.
65. Ren Y, Cheung HW, von Maltzhan G, et al. Targeted tumor-penetrating siRNA nanocomplexes for
credentialing the ovarian cancer oncogene ID4. Sci Transl Med. 2012;4:147ra12.
66. Davis ME, Zuckerman JE, Choi CHJ, et al. Evidence of RNAi in humans from systemically
administered siRNA via targeted nanoparticles. Nature. 2010;464:1067-70.
67. Engelberth SA, Hempel N, Bergkvist M. Development of nanoscale approaches for ovarian cancer
therapeutics and diagnostics. Crit Rev Oncog. 2014;19:281-315.
68. Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in
epithelial ovarian cancer. N Engl J Med. 2003;348:203-13.
69. Drerup JM, Liu Y, Padron AS, et al. Immunotherapy for ovarian cancer. Curr Treat Options Oncol.
2015;16:317.
70. Kandalaft LE, Powell DJ Jr, Coukos G. A phase I clinical trial of adoptive transfer of folate
receptor-alpha redirected autologous T cells for recurrent ovarian cancer. J Transl Med. 2012;10:157.
71. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of
genetically engineered lymphocytes. Science. 2006;314:126-9.
72. Vlad AM, Budiu RA, Lenzner DE, et al. A phase II trial of intraperitoneal interleukin-2 in patients
with platinum-resistant or platinum-refractory ovarian cancer. Cancer Immunol Immunother.
2010;59:293-301.
Source URL:
http://www.cancernetwork.com/oncology-journal/intraoperative-hyperthermic-intraperitoneal-chemot
herapy-patients-advanced-ovarian-cancer
Links:
[1] http://www.cancernetwork.com/review-article
[2] http://www.cancernetwork.com/oncology-journal
[3] http://www.cancernetwork.com/gynecologic-cancers
[4] http://www.cancernetwork.com/ovarian-cancer
[5] http://www.cancernetwork.com/authors/anton-oseledchyk-md
[6] http://www.cancernetwork.com/authors/oliver-zivanovic-md-phd
Page 11 of 11