REVIEW For reprint orders, please contact: [email protected]

Cytoreductive surgery for advanced ovarian cancer Martin Pölcher*1, Oliver Zivanovic2 & Dennis S Chi2 The amount of the largest diameter of visible residual tumor after cytoreductive surgery remains one of the strongest prognostic factors in advanced ovarian cancer. The implementation of a more aggressive surgical approach to increase the proportion of patients without visible residual tumor is, therefore, a rational concept. Thus, the surgical management of advanced ovarian, primary peritoneal and fallopian tube cancers now incorporates more comprehensive surgical procedures. However, these more extensive surgical procedures are associated with an increased risk of morbidity, which may have a negative impact on the oncologic outcome. In addition, it is unclear whether all patients benefit from a comprehensive surgical intervention in the same way or if there are patients whose disease course will not be influenced by this approach. The methodologic analysis of surgical effectiveness is complex and controversial owing to a lack of prospective surgical trials. This review acknowledges controversies and aims to discuss novel developments in the field of cytoreductive surgery for patients with ovarian, primary peritoneal and fallopian tube cancers. The focus of the review is to discuss the role of surgery at initial diagnosis. The role of secondary and tertiary surgery in the recurrent setting is beyond the scope of this review.

Ovarian, primary peritoneal and fallopian tube cancers are the most lethal gynecologic malignancies, with 22,280 estimated new cases and 15,500 estimated deaths in 2012 in the USA [1]. The majority of patients (60–70%) present at an advanced disease stage at the time of initial diagnosis with overt peritoneal extra­pelvic disease, lymph node involvement or distant metastatic disease beyond the peritoneal cavity and parenchymal liver metastasis (International Federation of Gynecology and Obstetrics [FIGO] stage III and IV, respectively) [1,2]. The natural history of advanced ovarian cancer is one of clinical remission after primary cytoreductive surgery (CRS) and post­operative chemo­ t herapy; however, the majority of patients recur within 24 months and long-term cure rates languish between 20 and 30%. Positive results from different improvement strategies have been reported over the past few years. Modification of the medical treatment after surgery demonstrated a benefit for patients treated with dose-dense intravenous (iv.) chemo­t herapy [3], for patients who underwent a combination of intraperitoneal (ip.) and iv. treatment [4], and for patients who were treated with a maintenance antiangio­genetic therapy after chemotherapy (mono­ c lonal anti-VEGF antibody, bevacizumab, or oral angio­genesis inhibitor, pazopanib [5–7]). Riskreducing strategies, for example, prophylactic salpingo-oophorectomy, reduce mortality in a 10.2217/WHE.14.4 © 2014 Future Medicine Ltd

high-risk population with germline mutations (BRCA1/2) [8]. However, effective screening strategies to detect early-stage diseases have not been established and continue to be elusive. Role of surgery

In contrast to other solid malignancies of the upper abdomen (e.g., pancreas, gallbladder and stomach), ovarian, primary peritoneal and fallo­pian tube carcinomas are sensitive to platinum-based combination chemotherapy in up to 80% of cases. The distinct chemo­sensitivity justifies a more extensive surgical approach. Surgical cytoreduction of advanced stage ovarian cancer, also termed ‘tumor debulking’, is defined as an attempt to maximally resect all visible and palpable disease. Postoperative residual disease is a variable of great importance for estimating the prognosis and guiding further treatment of affected patients. Radical surgical approach for advanced ovarian carcinoma

The term ‘radical surgery’ used for cancer surgery was introduced by William Halsted at the beginning of the 20th century and originates from the word radix, for example, removing the cancerous lesion including the root (radix) with tumor-free microscopic margins. In analogy, William Halsted surgically removed the entire breast, including the underlying muscle and the axillary lymph nodes to reduce the risk of Women's Health (2014) 10(2), 179–190

Red Cross Women’s Hospital Munich, Department of Gynecologic Oncology & Minimally-Invasive Surgery, Munich, Germany 2 Department of Surgery, Gynecology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA *Author for correspondence: [email protected] 1

Keywords • advanced ovarian carcinoma • cytoreductive surgery • hyperthermic intraoperative intraperitoneal chemotherapy

part of

ISSN 1745-5057

179

Review – Pölcher, Zivanovic & Chi local recurrence in patients with breast cancer. It was not until the late 1970s that Bernhard Fisher was able to prove equivalent outcomes for patients treated with less radical breastconserving surgery followed by local radiation therapy in patients with breast cancer. Unlike breast cancer, ovarian cancer involves the peritoneal surface with diffuse peritoneal carcinomatosis and the goal of CRS is to resect all macroscopic tumor lesions, not to remove the tumor with a tumor-free microscopic margin status. The goal of surgical cytoreduction of advanced stage ovarian cancer is to maximally resect all visible and palpable disease. There is a large body of evidence that demonstrates a survival benefit for patients who are left with minimal (‘optimally cytoreduced’) disease after surgery. For more than 30 years, it has been known that women with advanced ovarian cancer, who are left with an apparent small volume of residual disease following primary surgery and go on to receive systemic chemo­t herapy, have a superior outcome compared with patients who are left with a larger volume disease after initial surgery. In 1934, Meigs suggested removing as much tumor as possible to enhance the effectiveness of postoperative radiation therapy in patients with advanced ovarian cancer [9]. The radicality of this surgical approach is not related to the resection of predefined anatomic compartments but, rather, is based on multiple procedures individually targeting the organs involved with disease with the goal to achieve ‘optimal debulking’ status. These procedures include, but are not limited to, hysterectomy and salpingooophorectomy, peritonectomy with or without gastrointestinal surgery, lymph node dissection, omentectomy and upper abdominal surgery. In 1975, Griffiths hypothesized a thres­ hold – a maximum tumor diameter – that may remain after CRS independent of the number of tumor lesions without compromising the benefit of the surgical procedure [10]. Hoskins et al. were able to confirm this hypothesis [11]. Thus, patients who undergo suboptimal cytoreduction sustain the morbidity of a cytoreductive attempt without the associated survival benefit. Consequently, the Gynecologic Oncology Group defined a maximum tumor diameter of 1 cm or less as an optimal surgical outcome strongly associated with survival. In addition, multiple retrospective studies and meta-analyses have demonstrated and confirmed that the amount of residual disease after CRS inversely correlates with progression-free 180

www.futuremedicine.com

and overall survival (OS), and that increasing optimal cyto­reduction rates with more extensive and complex surgical efforts is associated with improved outcomes [12–18]. Prognostic impact of postoperative residual disease

Although, to date, the benefit of surgical cytoreduction has not been prospectively established through a well-defined prospective Phase III trial, CRS with the goal of achieving minimal residual disease in combination with platinum- and taxane-based chemotherapy is currently the accepted standard management. However, in the absence of prospective trials, we cannot say for sure whether the surgery itself is responsible for the superior outcome associated with small-volume residual disease or whether these patients are part of a biologically more favorable subgroup with excellent response to postoperative chemotherapy. The evidence strongly supporting the role of surgery has been derived from different types of ana ­lysis. Single-center studies

A retrospective analysis of 465 patients at Memorial Sloan-Kettering Cancer Center (NY, USA) with ‘bulky’ FIGO stage IIIC disease (those stage IIIC patients with nodal involvement only were excluded) revealed three distinct groups with significantly different survival rates (p 1 cm [13]. Although the difference in survival in patients left with smaller volume ≤1 cm was significant when compared with those with suboptimal residual disease, there was a trend in patients with gross up to 0.5 cm residual compared with those with 0.6 cm up to 1 cm (p = 0.06), suggesting that each incremental decrease in residual tumor below 1 cm may be associated with an incremental improvement in OS. Aletti et al. analyzed a patient cohort of 194 patients, including 144 with peritoneal carcinomatosis [12]. The only independent predictor of survival was residual disease of 1 cm, radical surgery in the abdominal cavity is debatable. Conclusion

The natural history of advanced ovarian cancer is one of clinical remission after primary CRS and postoperative chemotherapy; however, the majority of patients recur within 24 months, and longterm cure rates languish between 20 and 30%. Since the amount of the largest diameter of visible residual tumor after CRS remains one of the strongest prognostic factors, an aggressive surgical approach is taken to increase the proportion of patients without visible residual tumor. However, more extensive surgical procedures are associated with an increased risk of morbidity, which may have a negative impact on the oncologic outcome. One promising new treatment strategy is to combine surgical cytoreduction with hyperthermic intraoperative ip. chemotherapy (HIPEC). future science group

Cytoreductive surgery for advanced ovarian cancer –

Further study of HIPEC, and development of additional treatment strategies, are needed. Future perspective: the role of HIPEC

Since EOC remains confined to the peritoneal cavity for most of its natural history, numerous theoretical studies evaluated the use of ip. drug delivery as a management strategy [72–74]. The studies predicted a pharmacologic advantage for ip. therapy with improved tumor cell access, longer half-life in the peritoneal compartment, increased dose intensity and slow peritoneal clearance, while still reaching sufficient levels of systemic exposure for longer periods of time. Further preclinical data for platinum agents revealed that resistence to the drugs can be overcome by exposing the tumor cells to higher ip. concentrations. Animal studies confirmed that the peritoneal cavity can be exposed to substantially greater concentrations of cytotoxic

Review

agents [75–77]. Owing to the production of severe side effects, these concentrations are not realistically attainable within the systemic compartment. Based on these data, several Phase III clinical trials have explored the clinical utility of postoperative ip. therapy of small-volume residual disease following maximal CRS of advanced EOC [4,78,79]. In these trials, after postoperative recovery cisplatin-based ip. therapy was delivered through an ip. access device and compared with standard iv. chemotherapy. In the most recent of these trials (GOG-172), there was a statistically significant increase in the median PFS and OS in the cisplatin/paclitaxel ip. treatment compared with the iv. group, with a benefit of 6 and 16 months, respectively. These findings have prompted the National Cancer Institute (MD, USA) to make a clinical announcement in 2006; since then, postoperative ip. treatment in patients with minimal residual disease after

Table 2. Ongoing studies evaluating the role of hyperthermic intraperitoneal chemotherapy for patients with ovarian cancer. Disease status (trial design)

ClinicalTrials. Study gov identifier

Platinum-sensitive recurrence NCT01539785 (randomized)

Hyperthermic Intra-peritoneal Chemotherapy (HIPEC) in Ovarian Cancer Recurrence (HORSE)

Primary stage III or only NCT01709487 pleural stage IV ovarian carcinoma after neoadjuvant chemotherapy (nonrandomized)

Feasibility Study of HIPEC for Patients With Stage III or Only Pleural Stage IV Ovarian Carcinoma in First Line Therapy

Primary epithelial ovarian/ NCT01628380 tubal (FIGO stage IIIC) with complete or partial clinical response after three cycles of neoadjuvant chemotherapy (randomized)

Phase 3 Trial Evaluating Hyperthermic Intraperitoneal Chemotherapy in Upfront Treatment of Stage IIIC Epithelial Ovarian Cancer (CHORINE)

FIGO stage III ovarian cancer after primary/ neoadjuvant chemotherapy (randomized)

Interval debulking Surgery ± Hyperthermic Intraperitoneal Chemotherapy in Stage III Ovarian Cancer

NCT00426257

Platinum-sensitive recurrence NCT01767675 (randomized)

Outcomes After Secondary Cytoreductive Surgery With or Without Carboplatin Hyperthermic Intraperitoneal Chemotherapy (HIPEC) Followed by Systemic Combination Chemotherapy for Recurrent Platinum-Sensitive Ovarian, Fallopian Tube, or Primary Peritoneal Cancer

Recurrent ovarian, primary NCT01659554 peritoneal and fallopian tube carcinomas (nonrandomized)

A Phase II Combined Modality Protocol of Debulking Surgery With HIPEC Followed by Intraperitoneal Chemotherapy for the Treatment of Recurrent Ovarian, Primary Peritoneal & Fallopian Tube Cancers

Recurrent ovarian cancer after second-line chemotherapy (randomized)

Hyperthermic Intra-Peritoneal Chemotherapy (HIPEC) in Relapse Ovarian Cancer Treatment (CHIPOR)

NCT01376752

FIGO: International Federation of Gynecology and Obstetrics.

future science group

Women's Health (2014) 10(2)

185

Review – Pölcher, Zivanovic & Chi surgical cytoreduction is increasingly used in the USA and continues to be the subject of ongoing randomized trials with new regimens and new/additional drugs [80]. However, a substantial percentage of the patients (60%) were unable to complete six cycles of cisplatin/paclitaxel ip. chemotherapy, most commonly secondary to abdominal discomfort due to postoperative adhesion barriers and ip. catheter-related complications. Therefore, the European Organization for Research and Treatment of Cancer (Brussels, Belgium) and other international organizations are still unconvinced by available data and still consider this treatment experimental [81]. However, despite only 48% of the patients receiving three or fewer cycles of ip. therapy, the ip. group had significant improvement in median survival compared with the iv. group. An alternative to the postoperative local drug delivery is the intraoperative chemotherapy. For several reasons, there has been increasing interest in the surgical oncology community for combining surgical cytoreduction and hyperthermic ip. drug delivery (HIPEC) in the management of solid tumors whose natural history remains principally localized to the peritoneal cavity. The advantages are as follows: • By giving the chemotherapy intraoperatively, drug exposure is optimal secondary to direct contact with the cancer cells without the barriers of postoperative adhesions;

• Intraoperatively, the chemotherapy can be delivered under highly standardized procedures and the surgeon can guarantee optimal distribution of the chemotherapy, and dwell times can be controlled; • Hyperthermia has been shown to increase the cytotoxic effect of many chemotherapeutic agents by increasing DNA-crosslinking and increasing the tumor penetration [82–84]. A comprehensive review of HIPEC is beyond the scope of this review, but such publications are available [85–92]. This approach has been explored in the treatment of patients with peritoneal malignancies, including pseudomyxoma peritonei, gastric cancer and colorectal malignancies, suggesting a survival benefit using this approach. Unfortunately, there is essentially no evidence from prospective trials to objectively confirm any claimed benefits associated with this approach for patients with advanced EOC. The existing studies on HIPEC are mostly retro­spective in nature, heterogeneous with regards to combined inclusion of primary and recurrent disease, and lack robust dose-finding and pharmaco­k inetic data. Until evidencebased trials yield further data, it is reasonable to conclude that surgical cytoreduction with HIPEC offers a rational, but still investigative, approach in the management of EOC, the use of which should be employed under

Executive summary Background • Ovarian, primary peritoneal and fallopian tube cancers are the most lethal gynecologic malignancies. Prognostic impact of postoperative residual disease • Retrospective studies and meta-analyses have demonstrated and confirmed that the amount of residual disease after cytoreductive surgery (CRS) inversely correlates with progression-free and overall survival. • Increasing optimal cytoreduction rates with more extensive and complex surgical efforts is associated with improved outcomes. Obtaining a higher rate of primary optimal cytoreduction • Advances in surgical training, technique and perioperative care have allowed surgeons to maximize cytoreductive efforts, enabling them to increase the rate of optimal cytoreduction. The role of neoadjuvant chemotherapy • The rate of patients with no gross residual tumor was significantly higher after neoadjuvant chemotherapy when compared with patients who underwent primary CRS. However, the increase in complete gross resection rates after neoadjuvant chemotherapy does not translate into improved outcomes observed in patients who undergo primary CRS. Risks associated with surgical cytoreduction • Surgical cytoreduction is associated with a risk of morbidity and mortality. Single center data quantified the mortality rate after CRS in advanced ovarian cancer to be between 1.4 and 2.5%. • Risk factors for higher mortality in these studies are age, performance status and comorbidities, as well as emergency treatment compared with patients who were treated electively. Video-assisted thoracoscopic surgery • Patients with pleural effusions may benefit from a video-assisted thoracoscopic surgery for diagnosis and intrathoracic cytoreduction. Hyperthermic intraperitoneal drug delivery • Combining surgical cytoreduction and hyperthermic intraperitoneal drug delivery is a rational, but still investigative, approach in the management of epithelial ovarian cancer.

186

www.futuremedicine.com

future science group

Cytoreductive surgery for advanced ovarian cancer –

the umbrella of Institutional Review Boardapproved clinical trials. One randomized Phase II trial has been initiated recently at the Memorial Sloan-Kettering Cancer Center [93]. This is a randomized trial designed to compare the efficacy and safety of carboplatin at a dose of 800 mg/m2 administered as HIPEC after secondary CRS for recurrent platinum-sensitive EOC. Patients with minimal residual disease of 5 mm or less will be randomized 1:1 to CRS with HIPEC (arm A; n = 49) or CRS only (arm B; n = 49). Both arms will receive standard postoperative iv. chemotherapy. The primary objective is to determine the proportion of patients who are without evidence of Papers of special note have been highlighted as: • of interest •• of considerable interest 1.

Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J. Clin. 62(1), 10–29 (2012).

2.

Heintz AP, Odicino F, Maisonneuve P et al. Carcinoma of the ovary. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int. J. Gynecol. Obstet. 95(Suppl. 1), S161–S192 (2006).

3.

4.

••

5.

6.

7.

Katsumata N, Yasuda M, Takahashi F et al. Dose-dense paclitaxel once a week in combination with carboplatin every 3 weeks for advanced ovarian cancer: a Phase 3, open-label, randomised controlled trial. Lancet 374(9698), 1331–1338 (2009). Armstrong DK, Bundy B, Wenzel L et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 354(1), 34–43 (2006). These findings prompted the National Cancer Institute (MD, USA) to make a clinical announcement in 2006. Based on the results of these randomized Phase III trials, a combination of intravenous and intraperitoneal administration of chemotherapy conveys a significant survival benefit among women with optimally debulked epithelial ovarian cancer compared with intravenous administration alone. Burger RA, Brady MF, Bookman MA et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N. Engl. J. Med. 365(26), 2473–2483 (2011). Perren TJ, Swart AM, Pfisterer J et al. A Phase 3 trial of bevacizumab in ovarian cancer. N. Engl. J. Med. 365(26), 2484–2496 (2011). Du Bois A, Floquet A, Kim JW et al. Randomized, double-blind, Phase III trial of

future science group

disease progression at 24 months after CRS with or without HIPEC. Current ongoing clinical trials are listed in Table 2 . Financial & competing interests disclosure The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript. pazopanib versus placebo in women who have not progressed after first-line chemotherapy for advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (AEOC): results of an international intergroup trial (AGO-OVAR16) J. Clin. Oncol. (Suppl.), Abstract LBA5503 (2013).

References

8.

9.

Kauff ND, Domchek SM, Friebel TM et al. Risk-reducing salpingo-oophorectomy for the prevention of BRCA1- and BRCA2-associated breast and gynecologic cancer: a multicenter, prospective study. J. Clin. Oncol. 26(8), 1331–1337 (2008). Meigs JV. Tumors of the Female Pelvic Organs. Macmillan, NY, USA (1934).

a combined exploratory analysis of 3 prospectively randomized Phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d’Investigateurs Nationaux Pour les Etudes des Cancers de l’Ovaire (GINECO). Cancer 115(6), 1234–1244 (2009). 15. 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. 25(24), 3621–3627 (2007). 16. Bristow RE, Tomacruz RS, Armstrong DK,

Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J. Clin. Oncol. 20(5), 1248–1259 (2002).

10. Griffiths CT. Surgical resection of tumor bulk

in the primary treatment of ovarian carcinoma. Natl Cancer Inst. Monogr. 42, 101–104 (1975). 11. Hoskins WJ, McGuire WP, Brady MF et al.

The effect of diameter of largest residual disease on survival after primary cytoreductive surgery in patients with suboptimal residual epithelial ovarian carcinoma. Am. J. Obstet. Gynecol. 170(4), 974–979; discussion 979–980 (1994). 12. Aletti GD, Dowdy SC, Gostout BS et al.

Aggressive surgical effort and improved survival in advanced-stage ovarian cancer. Obstet. Gynecol. 107(1), 77–85 (2006). 13. 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. 103(2), 559–564 (2006). ••

Review

This study is important because it shows that, among those patients with larger residual disease, size does not affect prognosis appreciably.

14. Du Bois A, Reuss A, Pujade-Lauraine E,

Harter P, Ray-Coquard I, Pfisterer J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer:

Women's Health (2014) 10(2)

••

This meta-analysis shows that maximal cytoreduction was one of the most powerful determinants of cohort survival among patients with stage III or IV ovarian carcinoma.

17. Elattar A, Bryant A, Winter-Roach BA,

Hatem M, Naik R. Optimal primary surgical treatment for advanced epithelial ovarian cancer. Cochrane Database Syst. Rev. 8, CD007565 (2011). 18. Chi DS, Eisenhauer EL, Zivanovic O et al.

Improved progression-free and overall survival in advanced ovarian cancer as a result of a change in surgical paradigm. Gynecol. Oncol. 114(1), 26–31 (2009). 19. Eisenkop SM, Spirtos NM. What are the

current surgical objectives, strategies, and technical capabilities of gynecologic oncologists treating advanced epithelial ovarian cancer? Gynecol. Oncol. 82(3), 489–497 (2001). 20. Zivanovic O, Eisenhauer EL, Zhou Q et al.

The impact of bulky upper abdominal disease cephalad to the greater omentum on surgical

187

Review – Pölcher, Zivanovic & Chi outcome for stage IIIC epithelial ovarian, fallopian tube, and primary peritoneal cancer. Gynecol. Oncol. 108(2), 287–292 (2008).

of computed tomography predictors of suboptimal primary cytoreduction in patients with advanced ovarian cancer. J. Clin. Oncol. 25(4), 384–389 (2007).

21. Chi DS, Barakat RR. Aggressive surgery and

ovarian cancer. J. Clin. Oncol. 24(15), 2395–2396; author reply 2396–2397 (2006).

32. Bristow RE, Duska LR, Lambrou NC et al.

A model for predicting surgical outcome in patients with advanced ovarian carcinoma using computed tomography. Cancer 89(7), 1532–1540 (2000).

22. Chi DS, Musa F, Dao F et al. An analysis of

patients with bulky advanced stage ovarian, tubal, and peritoneal carcinoma treated with primary debulking surgery (PDS) during an identical time period as the randomized EORTC–NCIC trial of PDS vs neoadjuvant chemotherapy (NACT). Gynecol. Oncol. 124(1), 10–14 (2012).

33. Dowdy SC, Mullany SA, Brandt KR,

Huppert BJ, Cliby WA. The utility of computed tomography scans in predicting suboptimal cytoreductive surgery in women with advanced ovarian carcinoma. Cancer 101(2), 346–352 (2004).

23. Harter P, Muallem ZM, Buhrmann C et al.

Impact of a structured quality management program on surgical outcome in primary advanced ovarian cancer. Gynecol. Oncol. 121(3), 615–619 (2011).

34. Memarzadeh S, Lee SB, Berek JS,

Farias-Eisner R. CA125 levels are a weak predictor of optimal cytoreductive surgery in patients with advanced epithelial ovarian cancer. Int. J. Gynecol. Cancer 13(2), 120–124 (2003).

24. Peiretti M, Zanagnolo V, Aletti GD et al.

Role of maximal primary cytoreductive surgery in patients with advanced epithelial ovarian and tubal cancer: surgical and oncological outcomes. Single institution experience. Gynecol. Oncol. 119(2), 259–264 (2010).

35. Kehoe SM, Hook J, Nankivell M et al.

Chemotherapy or upfront surgery for newly diagnosed advanced ovarian cancer: results from the MRC CHORUS trial. Presented at: 2013 American Society of Clinical Oncology Annual Meeting. Chicago, IL, USA, May 31–June 4 2013.

25. Markman M. Concept of optimal surgical

cytoreduction in advanced ovarian cancer: a brief critique and a call for action. J. Clin. Oncol. 25, 4168–4170 (2007).

36. Vergote I, Tropé CG, Amant F et al.;

European Organization for Research and Treatment of Cancer Gynaecological Cancer Group, NCIC Clinical Trials Group. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 363(10), 943–953 (2010).

26. Covens AL. A critique of surgical

cytoreduction in advanced ovarian cancer. Gynecol. Oncol. 78(3 Pt 1), 269–274 (2000). 27. Chi DS, Franklin CC, Levine DA et al.

Improved optimal cytoreduction rates for stages IIIC and IV epithelial ovarian, fallopian tube, and primary peritoneal cancer: a change in surgical approach. Gynecol. Oncol. 94(3), 650–654 (2004). 28. Zivanovic O, Sima CS, Iasonos A et al.

The effect of primary cytoreduction on outcomes of patients with FIGO stage IIIC ovarian cancer stratified by the initial tumor burden in the upper abdomen cephalad to the greater omentum. Gynecol. Oncol. 116(3), 351–357 (2010). 29. Eisenhauer EL, D’Angelica MI,

Abu-Rustum NR et al. Incidence and management of pleural effusions after diaphragm peritonectomy or resection for advanced mullerian cancer. Gynecol. Oncol. 103, 871–877 (2006). 30. Aletti GD, Dowdy SC, Podratz KC,

Cliby WA. Surgical treatment of diaphragm disease correlates with improved survival in optimally debulked advanced stage ovarian cancer. Gynecol. Oncol. 100(2), 283–287 (2006). 31. Axtell AE, Lee MH, Bristow RE et al.

Multi-institutional reciprocal validation study

188



This is the first randomized trial exploring the role of neoadjuvant chemotherapy in patients with advanced ovarian cancer. The trial demonstrated no difference in progression-free and overall survival between primary surgery versus neoadjuvant chemotherapy.

37. Kommos S, Rochon J, Harter P et al.

Prognostic impact of additional extended surgical procedures in advanced stage primary ovarian cancer. Ann. Surg. Oncol 17, 279–286 (2010). 38. Colombo PE, Mourregot A, Fabbro M et al.

Aggressive surgical strategies in advanced ovarian cancer: a monocentric study of 203 stage IIIC and IV patients. Eur. J. Surg. Oncol. 35(2), 135–143 (2009). 39. Chekerov R, Braicu I, Castillo-Tong DC et al.

Outcome and clinical management of 275 patients with advanced ovarian cancer International Federation of Obstetrics and Gynecology II to IV inside the European Ovarian Cancer Translational Research Consortium-OVCAD. Int. J. Gynecol. Cancer 23(2), 268–275 (2013).

www.futuremedicine.com

40. Luyckx M, Leblanc E, Filleron T et al.

Maximal cytoreduction in patients with FIGO stage IIIC to stage IV ovarian, fallopian, and peritoneal cancer in day-to-day practice: a retrospective French multicentric study. Int. J. Gynecol. Cancer 22(8), 1337–1343 (2012). 41. Chi DS, Zivanovic O, Levinson KL et al.

The incidence of major complications after the performance of extensive upper abdominal surgical procedures during primary cytoreduction of advanced ovarian, tubal, and peritoneal carcinomas. Gynecol. Oncol. 119(1), 38–42 (2010). 42. Barlin JN, Yu C, Hill EK et al. Nomogram

for predicting 5-year disease-specific mortality after primary surgery for epithelial ovarian cancer. Gynecol. Oncol. 125(1), 25–30 (2012). 43. Fotopoulou C, Savvatis K,

Steinhagen-Thiessen E, Bahra M, Lichtenegger W, Sehouli J. Primary radical surgery in elderly patients with epithelial ovarian cancer: analysis of surgical outcome and long-term survival. Int. J. Gynecol. Cancer 20(1), 34–40 (2010). 44. Thrall MM, Goff BA, Symons RG, Flum DR,

Gray HJ. Thirty-day mortality after primary cytoreductive surgery for advanced ovarian cancer in the elderly. Obstet. Gynecol. 118(3), 537–547 (2011). 45. Wright JD, Lewin SN, Deutsch I et al.

Defining the limits of radical cytoreductive surgery for ovarian cancer. Gynecol. Oncol. 123(3), 467–473 (2011). 46. Wright JD, Herzog TJ, Neugut AI et al.

Effect of radical cytoreductive surgery on omission and delay of chemotherapy for advanced-stage ovarian cancer. Obstet. Gynecol. 120(4), 871–881 (2012). 47. Tebes SJ, Cardosi R, Hoffman MS. Colorectal

resection in patients with ovarian and primary peritoneal carcinoma. Am. J. Obstet. Gynecol. 195(2), 585–589; discussion 589–590 (2006). 48. Estes JM, Leath CA 3rd, Straughn JM Jr et al.

Bowel resection at the time of primary debulking for epithelial ovarian carcinoma: outcomes in patients treated with platinum and taxane-based chemotherapy. J. Am. Coll. Surg. 203(4), 527–532 (2006). 49. Peiretti M, Bristow RE, Zapardiel I et al.

Rectosigmoid resection at the time of primary cytoreduction for advanced ovarian cancer. A multi-center analysis of surgical and oncological outcomes. Gynecol. Oncol. 126(2), 220–223 (2012). 50. Gallotta V, Fanfani F, Vizzielli G et al.

Douglas peritonectomy compared to recto-sigmoid resection in optimally cytoreduced advanced ovarian cancer patients: analysis of morbidity and

future science group

Cytoreductive surgery for advanced ovarian cancer –

oncological outcome. Eur. J. Surg. Oncol. 37(12), 1085–1092 (2011). 51. Aletti GD, Podratz KC, Jones MB, Cliby WA.

Role of rectosigmoidectomy and stripping of pelvic peritoneum in outcomes of patients with advanced ovarian cancer. J. Am. Coll. Surg. 203(4), 521–526 (2006). 52. Bristow RE, Del Carmen MG, Kaufman HS,

Montz FJ. Radical oophorectomy with primary stapled colorectal anastomosis for resection of locally advanced epithelial ovarian cancer. J. Am. Coll. Surg. 197(4), 565–574 (2003). 53. Lim SW, Lim SB, Park JY, Park SY, Choi HS,

Jeong SY. Outcomes of colorectal anastomoses during pelvic exenteration for gynaecological malignancy. Br. J. Surg. 95(6), 770–773 (2008). 54. Devolder K, Amant F, Neven P, Van Gorp T,

Leunen K, Vergote I. Role of diaphragmatic surgery in 69 patients with ovarian carcinoma. Int. J. Gynecol. Cancer 18(2), 363–368 (2008). 55. Dowdy SC, Loewen RT, Aletti G, Feitoza SS,

Cliby W. Assessment of outcomes and morbidity following diaphragmatic peritonectomy for women with ovarian carcinoma. Gynecol. Oncol. 109(2), 303–307 (2008). 56. Gouy S, Chereau E, Custodio AS et al.

Surgical procedures and morbidities of diaphragmatic surgery in patients undergoing initial or interval debulking surgery for advanced-stage ovarian cancer. J. Am. Coll. Surg. 210(4), 509–514 (2010). 57. Tsolakidis D, Amant F, Van Gorp T,

Leunen K, Neven P, Vergote I. Diaphragmatic surgery during primary debulking in 89 patients with stage IIIB–IV epithelial ovarian cancer. Gynecol. Oncol. 116(3), 489–496 (2010). 58. Zapardiel I, Peiretti M, Zanagnolo V et al.

Diaphragmatic surgery during primary cytoreduction for advanced ovarian cancer: peritoneal stripping versus diaphragmatic resection. Int. J. Gynecol. Cancer 21(9), 1698–1703 (2011). 59. Fanfani F, Fagotti A, Gallotta V et al. Upper

abdominal surgery in advanced and recurrent ovarian cancer: role of diaphragmatic surgery. Gynecol. Oncol. 116(3), 497–501 (2010). 60. Chereau E, Rouzier R, Gouy S et al.

Morbidity of diaphragmatic surgery for advanced ovarian cancer: retrospective study of 148 cases. Eur. J. Surg. Oncol. 37(2), 175–180 (2011). 61. Matigbay PM, Adams PB, Silverman MB,

Cha SS, Podratz KC. Splenectomy as part of cytoreductive surgery in ovarian cancer. Gynecol. Oncol. 102, 369–374 (2006).

future science group

62. Eisenkop SM, Spirtos NM, Lin WC.

Splenectomy in the context of primary cytoreductive operations for advanced epithelial ovarian cancer. Gynecol. Oncol. 100(2), 344–348 (2006).

surgery (VATS) in patients with suspected advanced ovarian malignancies and pleural effusions. Gynecol. Oncol. 104(3), 670–674 (2007). 72. Dedrick RL. Theoretical and experimental

63. Kehoe SM, Abu-Rustum NR, Sonoda Y.

Incidence and management of pancreatic leaks after splenectomy with distal pancreatectomy performed during primary cytoreductive surgery for advanced ovarian cancer. Presented at: the 38th Annual Meeting on Women’s Cancer of the Society of Gynecologic Oncologists. San Diego, CA, USA, 3–7 March 2007. 64. Martinez A, Pomel C, Mery E, Querleu D,

Gladieff L, Ferron G. Celiac lymph node resection and porta hepatis disease resection in advanced or recurrent epithelial ovarian, fallopian tube, and primary peritoneal cancer. Gynecol. Oncol. 121(2), 258–263 (2011). 65. Raspagliesi F, Ditto A, Martinelli F,

Haeusler E, Lorusso D. Advanced ovarian cancer: omental bursa, lesser omentum, celiac, portal and triad nodes spread as cause of inaccurate evaluation of residual tumor. Gynecol. Oncol. 129(1), 92–96 (2013).

bases of intraperitoneal chemotherapy. Semin. Oncol. 12(3 Suppl. 4), 1–6 (1985). 73. Jones RB, Myers CE, Guarino AM,

Dedrick RL, Hubbard SM, Devita VT. High volume intraperitoneal chemotherapy (‘belly bath’) for ovarian cancer. Pharmacologic basis and early results. Cancer Chemother. Pharmacol. 1(3), 161–166 (1978). 74. Flessner MF, Dedrick RL, Rippe B.

Estimation of lymphatic absorption and intraperitoneal volume during hypertonic peritoneal dialysis. ASAIO Trans. 35(2), 178–181 (1989). 75. Los G, Mutsaers PH, Lenglet WJ, Baldew GS,

McVie JG. Platinum distribution in intraperitoneal tumors after intraperitoneal cisplatin treatment. Cancer Chemother. Pharmacol. 25(6), 389–394 (1990). 76. Los G, Ruevekamp M, Bosnie N,

De Graaf PW, McVie JG. Intraperitoneal tumor growth and chemotherapy in a rat model. Eur. J. Cancer Clin. Oncol. 25(12), 1857–1866 (1989).

66. Song YJ, Lim MC, Kang S et al. Extended

cytoreduction of tumor at the porta hepatis by an interdisciplinary team approach in patients with epithelial ovarian cancer. Gynecol. Oncol. 121(2), 253–257 (2011).

77. Los G, Verdegaal EM, Mutsaers PH,

McVie JG. Penetration of carboplatin and cisplatin into rat peritoneal tumor nodules after intraperitoneal chemotherapy. Cancer Chemother. Pharmacol. 28(3), 159–165 (1991).

67. Eisenkop SM, Spirtos NM. Procedures

required to accomplish complete cytoreduction of ovarian cancer: is there a correlation with ‘biological aggressiveness’ and survival? Gynecol. Oncol. 82(3), 435–441 (2001).

78. 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. 335(26), 1950–1955 (1996).

68. 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. 26(1), 83–89 (2008). 69. Chi DS, Abu-Rustum NR, Sonoda Y et al.

The benefit of video-assisted thoracoscopic surgery before planned abdominal exploration in patients with suspected advanced ovarian cancer and moderate to large pleural effusions. Gynecol. Oncol. 94(2), 307–311 (2004). 70. Chi DS, Barlin JN, Ramirez PT et al.

Follow-up study of the correlation between postoperative computed tomographic scan and primary surgeon assessment in patients with advanced ovarian, tubal, or peritoneal carcinoma reported to have undergone primary surgical cytoreduction to residual disease of 1 cm or smaller. Int. J. Gynecol. Cancer 20(3), 353–357 (2010). 71. Juretzka MM, Abu-Rustum NR, Sonoda Y

et al. The impact of video-assisted thoracic

Women's Health (2014) 10(2)

Review

••

This study revealed a statistically significant improvement in overall survival associated with regional delivery of cisplatin.

79. 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. 19(4), 1001–1007 (2001). 80. National Cancer Institute. Intraperitoneal

Therapy for Ovarian Cancer (2006). www.cancer.gov/clinicaltrials/conducting/ developments/ipchemo-digest/Page1

189

Review – Pölcher, Zivanovic & Chi 81. Gore M, Du Bois A, Vergote I. Intraperitoneal

chemotherapy in ovarian cancer remains experimental. J. Clin. Oncol. 24(28), 4528–4530 (2006). ••

Despite the National Cancer Institute announcement, the European Organization for Research and Treatment of Cancer (Brussels, Belgium) and the Arbeitsgemeinschaft Gynäkologische Onkologie-Germany (Munich, Germany) are unconvinced by available data, and still consider this treatment experimental, mainly because of the higher toxicity rates and the high rate of discontinuation.

82. Los G, Sminia P, Wondergem J et al.

Optimisation of intraperitoneal cisplatin therapy with regional hyperthermia in rats. Eur. J. Cancer 27(4), 472–477 (1991). 83. 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. 46(7), 1229–1237 (1993). 84. Los G, van Vugt MJ, Pinedo HM.

Response of peritoneal solid tumours after intraperitoneal chemohyperthermia treatment with cisplatin or carboplatin. Br. J. Cancer 69(2), 235–241 (1994).

190

85. Piso P, Dahlke MH, Ghali N et al.

Multimodality treatment of peritoneal carcinomatosis from colorectal cancer: first results of a new German centre for peritoneal surface malignancies. Int. J. Colorectal Dis. 22(11),1295–300 (2007). 86. Piso P, Dahlke MH, Loss M, Schlitt HJ.

Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in peritoneal carcinomatosis from ovarian cancer. World J. Surg. Oncol. 2, 21 (2004). 87. 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 113(2), 315–325 (2008). 88. Sugarbaker PH. Peritoneum as the first-line of

defense in carcinomatosis. J. Surg. Oncol. 95(2), 93–96 (2007). 89. Sugarbaker PH. Carcinomatosis – is cure an

option? J. Clin. Oncol. 21(5), 762–764 (2003). 90. Verwaal VJ, Bruin S, Boot H, van Slooten G,

van Tinteren H. 8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with

www.futuremedicine.com

peritoneal carcinomatosis of colorectal cancer. Ann. Surg. Oncol. 15(9), 2426–2432 (2008). 91. 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. 21(20), 3737–3743 (2003). 92. Yang XJ, Huang CQ, Suo T et al.

Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves survival of patients with peritoneal carcinomatosis from gastric cancer: final results of a Phase III randomized clinical trial. Ann. Surg. Oncol. 18(6), 1575–1581 (2011). 93. ClincialTrials.gov. A Phase II Combined

Modality Protocol of Debulking Surgery With HIPEC Followed by Intraperitoneal Chemotherapy for the Treatment of Recurrent Ovarian, Primary Peritoneal & Fallopian Tube Cancers. http://clinicaltrials.gov/show/NCT01659554 94. Aletti GD, Dowdy SC, Gostout BS et al.

Quality improvement in the surgical approach to advanced ovarian cancer: the Mayo Clinic experience. J. Am. Coll. Surg. 208(4), 614–620 (2009).

future science group

Cytoreductive surgery for advanced ovarian cancer.

The amount of the largest diameter of visible residual tumor after cytoreductive surgery remains one of the strongest prognostic factors in advanced o...
770KB Sizes 5 Downloads 3 Views