28
Hyperthermia in Cancer Therapy Page 1 of 28 http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015 Aetna Better Health® 2000 Market Street, Suite 850 Philadelphia, PA 19103 AETNA BETTER HEALTH® Clinical Policy Bulletin: Hyperthermia in Cancer Therapy Number: 0278 Policy I. Aetna considers the following procedures medically necessary: A. Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy for the treatment of pseudomyxoma peritonei. B. Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy for the treatment of peritoneal mesothelioma. C. Regional hyperthermic melphalan perfusion in members with stage II, IIIA, and stage III in-transit extremity melanoma. D. Sequential radiation -- local/regional external hyperthermia only for superficial recurrent melanoma, locally advanced/recurrent breast cancers and cervical lymph node metastases from head and neck cancer. II. Aetna considers hyperthermia experimental and investigational for all other indications including the following applications because of insufficient evidence regarding its effectiveness in these conditions: A. Deep hyperthermia alone or in combination with radiation therapy. B. Hyperthermic intrapleural chemotherapy for intrapleural mesothelioma. C. Hyperthermic administration of intraperitoneal chemotherapy for appendiceal carcinoma without pseudomyxoma, bladder cancer, clear cell carcinoma of the ovary, colon cancer, colorectal signet ring carcinoma, desmoplastic small round cell tumor, gastric cancer, goblet carcinoid tumor, hepatocellular carcinoma, ovarian cancer, pancreatic cancer, small bowel adenocarcinoma, thymic carcinoma, or uterine leiomyosarcoma. D. Regional hyperthermic melphalan perfusion in stage I, IIIB and IIIAB extremity melanoma, as well as regional hyperthermic perfusion for extremity melanoma in conjunction with any other chemotherapy. E. Interstitial, intra-cavitary, and intraluminal hyperthermia. F. Whole body hyperthermia for testicular cancer and other indications.

Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Embed Size (px)

Citation preview

Page 1: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 1 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

Aetna Better Health® 2000 Market Street, Suite 850 Philadelphia, PA 19103

AETNA BETTER HEALTH®

Clinical Policy Bulletin: Hyperthermia in Cancer Therapy

Number: 0278

Policy

I. Aetna considers the following procedures medically necessary:

A. Cytoreductive surgery combined with hyperthermic intraperitoneal

chemotherapy for the treatment of pseudomyxoma peritonei.

B. Cytoreductive surgery combined with hyperthermic intraperitoneal

chemotherapy for the treatment of peritoneal mesothelioma.

C. Regional hyperthermic melphalan perfusion in members with stage

II, IIIA, and stage III in-transit extremity melanoma.

D. Sequential radiation -- local/regional external hyperthermia only for

superficial recurrent melanoma, locally advanced/recurrent breast

cancers and cervical lymph node metastases from head and neck

cancer.

II. Aetna considers hyperthermia experimental and investigational for all other

indications including the following applications because of insufficient

evidence regarding its effectiveness in these conditions:

A. Deep hyperthermia alone or in combination with radiation therapy.

B. Hyperthermic intrapleural chemotherapy for intrapleural

mesothelioma.

C. Hyperthermic administration of intraperitoneal chemotherapy

for appendiceal carcinoma without pseudomyxoma, bladder cancer,

clear cell carcinoma of the ovary, colon cancer, colorectal signet ring

carcinoma, desmoplastic small round cell tumor, gastric cancer,

goblet carcinoid tumor, hepatocellular carcinoma, ovarian

cancer, pancreatic cancer, small bowel adenocarcinoma, thymic

carcinoma, or uterine leiomyosarcoma.

D. Regional hyperthermic melphalan perfusion in stage I, IIIB and IIIAB

extremity melanoma, as well as regional hyperthermic perfusion for

extremity melanoma in conjunction with any other chemotherapy.

E. Interstitial, intra-cavitary, and intraluminal hyperthermia.

F. Whole body hyperthermia for testicular cancer and other indications.

Page 2: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 2 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

G. Intraperitoneal hyperthermic chemotherapy for peritoneal surface

malignancy (peritoneal carcinomatosis) for indications other than

pseudomyxoma peritonei or peritoneal mesothelioma.

H. Regional hyperthermic perfusion for indications (e.g., non-small cell

lung cancer) other than extremity melanoma.

I. Superficial hyperthermia for paranasal sinus and nasal cavity

cancer.

J. Transrectal ultrasound hyperthermia for prostate cancer.

Note: Stages of melanoma (0 to IV) can be found in the following link:

http://www.cancer.org/cancer/skincancer-melanoma/detailedguide/melanoma-skin

-cancer-staging

Background

Several studies support the hypothesis that hyperthermia appears to potentiate the

tumoricidal effects of radiation therapy. Radiation therapy and hyperthermia

probably act independently. Studies have suggested a potential synergistic

response to radiation therapy and hyperthermia versus radiation therapy alone.

The goal of hyperthermia in cancer therapy is to produce tumor tissue

temperatures above 41 to 42 degrees centigrade. Above this temperature, heat

has a direct cytotoxic effect on both normal and tumor cells and also has a

radiosensitizing effect by preventing repair of sublethal and potentially lethal

radiation damage. Heat can also potentiate the cytotoxic effect of a variety of

chemotherapeutic agents.

There are several different heating systems used. The 3 physical modalities

employed for power deposition in local and regional clinical hyperthermia are (i)

ultrasound, (ii) electromagnetic fields, and (iii) electromagnetic radiation. In

addition, there are 3 methods of inducing hyperthermia interstitially: (i)

radiofrequency needle electrodes, (ii) coaxial microwave antennas, and (iii) hot

sources.

Hyperthermia has been shown to potentiate the effect of radiation therapy in the

treatment of superficial lesions (less than 3 cm in depth). Clinical experience has

largely been limited to treatment of recurrent, metastatic superficial melanomas,

chest wall recurrence of breast cancer and cervical lymph node metastases from

head and neck cancers. Tumor depth is a critical factor when combining radiation

therapy and hyperthermia. Lesions less than 3 cm from the surface treated with

radiation therapy and hyperthermia have been shown to have a significantly

greater complete response rate compared to the complete reponse rate of lesions

greater than 3 cm deep.

Guidelines on breast cancer from the National Comprehensive Cancer Network

(NCCN, 2009) include consideration of the addition of hyperthermia to irradiation

for localized recurrences/metastastes. The NCCN guidelines explain that there

have been several prospective randomized trials comparing radiation to radiation

plus hyperthermia in the treatment of locally advanced/recurrent cancers, primarily

breast cancer chest wall recurrences (Vernon et al, 1996; Jones et al, 2005). The

Page 3: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 3 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

NCCN guidelines state that, while there is heterogeneity among the study results,

a recent series with strict quality assurance demontrated a statistically significant

increase in local tumor response and greater duration of local control with the

addition of hyperthermia to radiation compared to radiation alone (Jones et al,

2005). However, no differences in overall survival have been demonstrated. The

NCCN guidelines state that delivery of local hyperthermia is technically demanding

and requires specialized expertise and equipment (e.g., the monitoring of

temperatures and management of possible tissue burns). The NCCN Panel thus

recommended the use of hyperthermia be limited to treatment centers with

appropriate training, expertise and equipment. The NCCN guidelines noted that

the addition of hyperthermia generated substantial discussion and controversy

among the NCCN panel members and is a category 3 recommendation (the

recommendation is based upon any level of evidence but reflects major

disagreement). The NCCN's clinical practice guideline on melanoma (version

2.2013) recommends the use of hyperthermic perfusion/infusion with melphalan

for "stage III in-transit" melanoma (category 2A recommendation).

The results of clinical studies combining hyperthermia and radiotherapy in the

treatment of melanoma have been complex and somewhat conflicting (e.g.,

Shidnia et al, 1990; Engin et al, 1993; Overgaard et al, 1995). A randomized trial

of radiotherapy with or without external hyperthermia was conducted by the

European Society for Hyperthermic Oncology in 70 patients with metastatic or

recurrent melanoma (Overgaard et al, 1995). The addition of hyperthermia to

radiotherapy increased the complete response rate from 35 % to 62 %, and 2-year

local control rates from 28 % to 46 %. However, only 14 % of patients received

the prescribed protocol treatment because of "equipment difficulties", and survival

did not differ between the 2 groups.

Hyperthermia plus chemotherapy appears to result in increased cell toxicity due to

net increase in DNA damage after exposure to hyperthermia and chemotherapy.

Regional hyperthermic melphalan isolated limb perfusion for stage II and IIIA

extremity melanoma has become routine practice. Isloated hyperthermic

mephalan limb perfusion of state II and IIIA extremity melanomas has been shown

to be effective in prospective, randomized trials. Surgery plus regional

hyperthermic melphalan perfusion versus surgery alone decreased recurrences

and improved survival significantly in several randomized studies. Also, a

comparison of responses after melphalan hyperthermic therapeutic limb perfusion

versus melphalan normothermic limb perfusion for extremity melanoma showed a

clear advantage in response rates. Although hyperthermic perfusion of stage IIIB

and IIIAB extremity melanoma shows a trend toward improvement in survival, well

controlled trials will be necessary to document the effectiveness of this technique

in node- positive patients. Similarly, while adjuvant treatment of stage I extremity

melanoma shows a trend toward improved 5-year survival, well-controlled studies

needed to confirm this observation.

Local or systemic hyperthermia alone has not been shown to be an effective

cancer treatment. Although interstitial radiation-hyperthermia appears promising,

well controlled trials comparing radiation therapy alone versus combined interstitial

radiation-hyperthermia are needed. Whole body heating is a complex, labor-

intensive technique that is difficult to accomplish and clinically impractical.

Page 4: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 4 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

Emami et al (1996) reported on a randomized trial that found no benefit to

interstitial radiation-hyperthermia in comparison with interstitial radiotherapy

alone on tumor regression or control in accessible lesions. From January 1986 to

June 1992, 184 patients with persistent or recurrent tumors after previous

radiotherapy and/or surgery, which were amenable to interstitial radiotherapy,

were accessioned to a protocol developed by the Radiation Therapy Oncology

Group (RTOG). A total of 173 cases were analyzed (87 patients in the interstitial

radiotherapy group and 86 in the interstitial radiation-hyperthermia arm). The

investigators stated that the 2 arms were well-balanced regarding stratification

criteria. Most tumors were in the head and neck (40 % in the interstitial radiation

therapy group and 46 % in the interstital radiation-hyperthermia group), and pelvis

(42 % and 43 %, respectively). Eighty-four percent of patients in both arms had

prior radiation therapy (40 Gy or more); 50 % and 40 %, respectively, had prior

surgery, and 34 % in each arm had prior chemotherapy. The investigators said

that the dose of radiation therapy administered was dependent on the previous

radiation dose and did not exceed a total cumulative dose of 100 Gy.

Hyperthermia was delivered in 1 or 2 sessions, either before or before and after

interstitial implant. According to the investigators, the intended goal of the

hyperthermia was to maintain a minimal tumor temperature of 42.5 degrees C for

30 to 60 mins. The investigators reported that there was no difference in any of

the study end points between the 2 arms. Complete response was 53 % and 55

% in both arms. Two-year survival was 34 % and 35 %, respectively. Complete

response rate for persistent lesions was 69 % and 63 % in the 2 treatment arms as

compared with 40 % and 48 % for recurrent lesions. The investigators reported

that a set of minimal adequacy criteria for the delivery of hyperthermia was

developed. When these criteria were applied, only 1 patient had an adequate

hyperthermia session. Acute grade 3 and 4 toxicities were 12 % for interstitial

radiation therapy and 22 % for insterstitial radiation-hyperthermia therapy. The

investigators reported that late grade 3 and 4 toxicities were 15 % for interstitial

radiation therapy and 20 % for interstitial hyperthermia radiation therapy. The

difference was not significant. The investigators concluded that interstitial

hyperthermia, as applied in this randomized study, did not show any additional

beneficial effects over interstitial radiotherapy alone. The investigators stated that

delivery of hyperthermia remains a major obstacle (since only 1 patient met the

basic minimum adequacy criteria as defined in this study). The investigators

stated that the benefit of hyperthermia in addition to radiation therapy still remains

to be proven in properly randomized prospective clinical trials after substantial

technical improvements in heat delivery and dosimetry are achieved.

In a multi-institutional, prospective, randomized trial sponsored by the International

Atomic Energy Agency, Mitsumori et al (2007) examined if the combination of

hyperthermia (HT) and radiotherapy (RT) improves the local response rate of

locally advanced non-small cell lung cancer (NSCLC) compared with that obtained

by RT alone. A total of 80 patients with locally advanced NSCLC were

randomized to receive either RT alone or radiation RT plus HT. The primary

endpoint was the local response rate. The secondary endpoints were local

progression-free survival (PFS) and overall survival (OS). The median follow-up

period was 204 days for all patients and 450 days for surviving patients. There

were no significant differences between the 2 arms with regard to local response

rate (p = 0.49) or OS rate (p = 0.868). However, local PFS was significantly better

Page 5: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 5 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

in the RT plus HT arm (p = 0.036). Toxicity was generally mild and no grade 3 late

toxicity was observed in either arm. The authors concluded that although

improvement of local PFS was observed in the RT plus HT arm, this prospective

randomized study failed to show any substantial benefit from the addition of HT to

RT in the treatment of locally advanced NSCLC.

In a phase II clinical study, Maluta et al (2007) evaluated feasibility and results in

terms of biochemical PFS, OS, and treatment toxicity profile of HT combined with

RT in locally advanced high risk prostate cancer. A total of 144 patients with

locally advanced prostate cancer (LAPC) were enrolled in this study. They were

treated using conformal RT (CRT) plus local HT (LHT) and androgen suppression

therapy (AST). Treatment modalities consisted of: (i) CRT with a mean dose of 74

Gy (2 Gy/fraction/5 fractions per week); (ii) LHT: 1 session per week during the

1st, 2nd, 3rd, and 4th week of the RT course; (iii) AST was administered as neo-

adjuvant and adjuvant therapy in more than 60 % of patients. The median follow-

up time was 51.7 months. Four patients were lost at follow-up. Of 140 evaluated

patients, 4 died because of inter-current diseases and 12 because of progression

of disease. Patients were evaluated in terms of 5-year OS (87 %), and 5-year

biochemical PFS (49 %). No significant side effects, except symptoms related to

AST have been reported. No late grade 3 toxicity occurred. The authors

concluded that in advanced high-risk prostatic cancer, HT is feasible and well-

tolerated. It may be useful to enhance the RT efficacy at intermediate dose in

order to avoid higher doses of irradiation which increases acute and late

sequelae. The advantage of LHT combined with CRT should be confirmed by a

randomized phase III trial, comparing irradiation plus AST with or without HT.

A number of studies have evaluated the use of hyperthermia combined with

chemotherapy for cervical cancer. A multi-institutional prospective randomized

controlled trial sponsored by the International Atomic Energy Agency found no

benefit to radiotherapy in combination with regional hyperthermia over

radiotherapy alone in rate of local control of cervical cancer (Vasanthan et al,

2005). A total of 110 patients with biopsy-proven, locally advanced

cervical carcinoma were randomized to treatment by radiotherapy with or without

hyperthermia. The patients were stratified by institution, stage, and histologic

type. Each patient received external beam radiation therapy and brachytherapy.

For the patients randomized to receive hyperthermia, a minimum of 5 sessions

(60 mins each, once per week) were administered, employing a radiofrequency

(RF) capacitive heating device. The median follow-up period was 466 days for all

the patients and 512 days for the surviving patients. The authors stated that the 2

arms were well-balanced with regard to the patient factors, tumor factors, and

treatment factors. The OS rate at 3 years was 73.2 %, and the local control rate

was 68.5 %. There were no significant differences between the patients treated

with or without hyperthermia, either with regard to the survival (p = 0.1893) or the

rate of local control (p = 0.58). The authors reported that survival was significantly

worse among the patients with stage IIb disease who received hyperthermia (p =

0.0162) although there was no difference in their rate of local control (p = 0.7988).

The authors explained that further analysis is necessary to determine if the

difference in survival is due to a greater incidence of distant metastases or some

other cause. Acute grade 2 to 3 toxicity was seen in 10 of 55 patients (18 %)

treated by hyperthermia and in 2 of 55 of the patients (4 %) treated without

hyperthermia (p = 0.01). There authors reported that there was no significant

Page 6: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 6 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

difference in the late toxicity observed in the 2 arms. The authors concluded that

this prospective randomized study failed to show any benefit from the addition of

hyperthermia to radiotherapy in the treatment of locally advanced cervical cancer.

The authors found that acute toxicity was significantly greater among the patients

receiving hyperthermia, and the survival was significantly worse among the stage

IIb patients receiving hyperthermia even though there was no difference in the

local control rate.

In a phase II study, Richel et al (2004) examined the efficacy and toxicity of whole-

body hyperthermia with carboplatin chemotherapy in persons with metastatic or

recurrent cervical cancer. Twenty-one of 25 participants were evaluable for

response: 1 complete remission, 6 partial responses, stable disease in 9 patients

and progression in 5, leading to a response rate of 33 %. The investigators

reported that 3 of 4 evaluable chemotherapy pre-treated patients progressed,

while this was seen in only 2 of 17 chemotherapy-naive patients. The median

survival is 7.8 months (range of 1.3 to 43+) and no patients were lost to follow-up.

The investigators reported that grades 3/4 toxicities were common: leukopenia in

35 %, thrombopenia in 61 % and anemia in 22 % of all treatments. The

investigators stated that excessive, partly reversible renal toxicity was seen in 2

patients (grades 3 and 4). The investigators concluded that the efficacy of whole

body hyperthermia and carboplatin in recurrent and/or metastatic cervical cancer

seems comparable to that of other palliative chemotherapy regimens in this

disease. The investigators stated that the considerable toxicity, though largely

manageable, includes unexpected and severe unacceptable renal toxicity, and

that this regimen seems less suitable for palliative care.

The Dutch Deep Hyperthermia Trial showed that combining radiotherapy with

hyperthermia impoved local control rates and survival over radiotherapy alone in

women with locally advanced cervical carcinoma (Van der Zee et al, 2000;

Franckena et al, 2008). It is not known, however, whether radiotherapy and

hyperthermia results in superior outcomes to radiotherapy and chemotherapy,

which is the current standard of care for advanced cervical carcinoma. Franckena

et al (2008) reported on the long-term results of the Dutch

Deep Hyperthermia Trial after 12 years of follow-up. From 1990 to 1996, a total of

114 women with locoregionally advanced cervical carcinoma were randomly

assigned to RT or RT+HT. The RT was applied to a median total dose of 68 Gy.

The HT was given once-weekly. The primary end point was local control.

Secondary end points were OS and late toxicity. At the 12-year follow-up, local

control remained better in the RT+HT group (37 % versus 56 %; p = 0.01). The

authors reported that survival was persistently better after 12 years: 20 % (RT)

and 37 % (RT+HT; p = 0.03). Grade 3 or higher radiation-induced late toxicities

were reported to be similar in both groups.

Harima et al (2001) reported on a small (n = 40) randomized controlled clinical trial

of RT versus RT+HT in persons with advanced cervical carcinoma, and found

improvements in response and 3-year local relapse-free survival with the addition

of HT. A total of 40 patients with stage IIIb cervical carcinoma were treated with

external beam radiotherapy to the pelvis, combined with iridium 192 high-dose-

rate intracavitary brachytherapy. Patients were randomly assigned to radiotherapy

alone or radiotherapy plus 3 sessions of hyperthermia. The investigators reported

that a complete response was achieved in 50 % of patients treated with RT versus

Page 7: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 7 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

80 % in patients receiving radiotherapy plus hyperthermia. There was also a trend

toward improvement in 3-year OS and disease-free survival (DFS) among patients

who were treated with RT+HT (58.2 % and 63.6 %) versus RT alone (48.1 % and

45 %), but the differences were not statistically significant. The authors reported

that the 3-year local relapse-free suvival of the patients who were treated with

RT+HT (79.7 %) was significantly better than that of the patients treated with RT

(48.5 %) (p = 0.048). The authors noted that RT+HT did not significantly add to

toxicity over RT alone.

Current guidelines recommend the use of chemotherapy plus radiotherapy

for advanced cervical cancer (e.g., NCCN, 2009; NCI, 2008). Although it has been

noted that the addition of hyperthermia to radiotherapy appears to improve

response rates and 3-year survival by a magnitude similar to that observed in

chemoradiotherapy studies in cervical cancer, Westermann et al (2005) has noted

that the reported randomized clinical trial experience of chemotherapy studies (n =

2,192 patients) far exceeds that for radiation therapy plus hyperthermia (n = 154

patients). Ongoing studies are examining the potential benefits of adding

hyperthermia to radiotherapy and chemotherapy in advanced cervical carcinoma

(Westermann et al, 2005).

A number of studies have examined the use of cytoreductive surgery

plus hyperthermic intraperitoneal chemotherapy as a treatment for pseudomyxoma

peritonei, a rare cancer arising from low-grade adenocarcinomas of appendiceal,

ovarian, or peritoneal origin. Reported outcomes of cytoreductive surgery

(CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) for this rare

condition are markedly superior to that reported for cytoreduction alone and

other conventional treatments for this condition (Sugarbaker and Chang, 1999;

Stewart et al, 2005; Elias et al, 2008).

Although some authorities (Esquivel et al, 2007) have recommended use

of HIPEC for patients with recurrent and/or metastatic colon caner with peritoneal

involvement, this recommendation is based upon uncontrolled studies. There are

a lack of phase III studies demonstrating improved outcomes with the use of

hyperthermia compared to non-hyperthermic administration. The sole randomized

controlled clinical trial randomly assigned 105 patients with peritoneal

carcinomatosis from appendiceal (n = 18) or colorectal (n = 87) cancer to either

standard treatment with systemic chemotherapy (fluorouracil-leucovorin) with or

without palliative surgery, or aggressive cytoreduction (debulking) and

hyperthermic intraperitoneal chemotherapy followed by the same systemic

chemotherapy regime (Verwaal et al, 2003). After a median follow-up period of 22

months, the median survival was 12.6 months in the standard therapy arm and 22

months in the hyperthermic intraperitoneal chemotherapy arm. The study has

been criticized because the HIPEC group differed from the standard therapy group

not only in the use of intraperitoneal hyperthermic chemotherapy, but also in

surgical debulking. It is possible that the important treatment was the surgical

debulking, and the HIPEC made little difference.

Guidelines from the National Comprehensive Cancer Network (2009) state that the

NCCN colon cancer guidelines panel considers the treatment of disseminated

carcinomatosis with cytoreductive surgery (i.e., peritoneal stripping surgery) and

Page 8: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 8 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

peri-operative HIPEC to be investigational and does not endorse such therapy

outside of a clinical trial.

Minicozzi and co-workers (2008) estimated the post-operative morbidity and

mortality and short-term outcome of treatment of the peritoneal carcinomatosis

(PC) by CRS and HIPEC. A total of 24 patients with PC or positive cytology at

peritoneal washing were treated. Primary tumor was ovarian carcinoma in 10

patients: 4 cases presented peritoneal surface malignancies (PSM) after any time

from hysteroadnexectomy related to primary tumor, 6 cases synchronous PSM.

Primary tumor was gastric cancer in 7 patients: the peritoneal washing was

positive in 4 cases and, during follow-up following gastrectomy, another 2 cases

presented PSM. One patient was previously treated with ovariectomy for ovarian

mass that resulted a Krukenberg's tumor of gastric cancer. Primary tumor was

pseudomixoma peritonei (PMP) in 4 patients; CRS and HIPEC was performed as

1st-line therapy in only 1 patient. Three patients were previously treated for colon

carcinoma; HIPEC was performed via the abdomino-pelvic cavity for 60 mins

using a closed abdomen technique. The drugs used were mitomycin C (3.3

mg/m2/L) and cisplatin (25 mg/m2/L). The intra-cavitary mean temperature was

41.8 degrees C. The mean peritoneal cancer index was 14. Post-operative major

complications occurred in 7 cases (28 %), post-operative minor complications

occurred in 8 cases (32 %). No patients died in the post-operative period. Mean

hospital stay was 11.5 days (6 to 35 days). After a median follow-up of 8 months

(range of 2 to 34), 14 (58 %) patients were alive and 13 were disease-free. The

authors concluded that these findings are consistent with other studies for the high

rate of post-operative morbidity associated with treatment, but they achieved best

results on mortality and post-operative hospital stay. The authors concluded

that CRS associated with HIPEC is a good therapeutic option especially in ovarian

-related carcinosis and PMP.

Scaringi et al (2008) evaluated the role of HIPEC, associated or not to CRS in the

treatment of different stages of advanced gastric cancer (AGC). A total of 37

patients with AGC who underwent 43 HIPEC were included in this study.

Hyperthermic intraperitoneal chemotherapy used mitomycin-C and cisplatin for 60

to 90 mins at 41 to 43 degrees C intra-abdominal temperature. The main

endpoints were long-term survivals, morbidity and mortality rates. Eleven patients

had no demonstrable sign of PC and constituted the prophylactic-group, while 26

patients had macroscopic PC (PC-group). Five patients were Gilly 1 or 2 (nodules

less than 0.5 cm) and 21 Gilly 3 or 4 (nodules greater than or equal to 0.5 cm). In

the PC-group a complete curative CRS was achieved before HIPEC in 8 (PC-

curative subgroup) and a palliative HIPEC in 18 patients (PC-palliative subgroup).

The overall 30-days mortality was 5 % (2 patients). Two patients in the

prophylactic group died within 6 months after hospital discharge (overall mortality

11 %). The estimated risk of death per procedure was 9 %. Ten patients (27 %)

presented 1 or more complications. The median survival was 23.4 months in the

prophylactic group, and 6.6 months in the PC-group (p < 0.05). The median

survival in the PC-curative subgroup was 15 versus 3.9 months in the PC-palliative

subgroup (p = 0.007). The median survival according to Gilly classification was

significantly different (Gilly 1 and 2 versus Gilly 3 and 4, 15 versus 4 months,

respectively, p = 0.014). The global recurrence rates between the prophylactic

group and the PC-curative subgroup at 2 years were 36 % versus 50 %,

respectively. The median delay to recurrence was 18.5 versus 9.7 months,

Page 9: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 9 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

respectively. The authors concluded that HIPEC might be useful to improve the

survival in selected patients with AGC only when a complete CRS can be

achieved. Despite encouraging data, prospective studies, based on larger cohorts

of patients are needed to evaluate the role of this procedure as a prophylactic

treatment in patients with AGC.

Spiliotis et al (2008) reported their experience in the combined treatment of PC

using CRS plus HIPEC. This prospective study included patients with PC from

gynecological, gastric and colon cancer, treated in 2 centers. Cytoreductive

surgery included the peritonectomy procedures described by Jacquet and

Sugarbaker as well as multi-visceral resections in order to achieve a complete

macroscopical cancer eradication. The HIPEC that followed was performed via

the open abdomen technique. A total of 24 patients (3 men and 21 women, mean

age of 60 years) were treated. Twelve patients had PC from ovarian cancer, 7

from colon, 3 from gastric and 2 from uterine cancer. The mean duration of the

procedure was 7.83 hours (range of 5 to 12.30). Macroscopically, complete

cytoreduction (CC) was achieved in 18 (75 %) patients. Two (8.3 %) patients died

in the first 30 days. The overall morbidity was 42 % and 2 patients were re-

operated. The mean follow-up was 22 months (range of 3 to 36). The overall 1-

year survival was 59.1 %; concerning the gynecological cancers it was 53.8 %

(mean survival of 11.7 months) and for gastrointestinal cancers it was 44.4 %

(mean survival of 9.5 months). The authors concluded that their findings

suggested that the combined treatment of CRS plus HIPEC for PC is associated

with acceptable mortality and morbidity and offers an improved survival in these

patients.

Di Giorgio et al (2008) examined the use of CRS (peritonectomy procedures)

combined with HIPEC in the treatment of diffuse PC from ovarian cancer. A total

of 47 patients with primary advanced or recurrent ovarian cancer and diffuse PC

were enrolled; 22 underwent primary and 25 secondary CRS plus immediate

HIPEC followed by systemic chemotherapy. The overall mean Sugarbaker

peritoneal cancer index was 14.9 (range of 6 to 28). A mean of 6 surgical

procedures were required per patient (range of 4 to 10). In 87.3 % of the patients

debulking achieved optimal cytoreduction (Sugarbaker completeness of

cytoreduction [CC] score 0 to 1), whereas in 12.7 % it left macroscopic residual

disease (CC-2 or CC-3). Major complications developed in 21.3 % of the patients

and the in-hospital mortality rate was 4.2 %. The mean OS was 30.4 months,

median survival was 24 months, and mean DFS was 27.4 months. Five-year

survival was 16.7 %. Uni-variate (log-rank test and analysis of variance) and multi

-variate analyses (Cox proportional-hazard model) identified the CC score as the

main factor capable of independently influencing survival. The authors concluded

that peritonectomy procedures combined with HIPEC offer promising long-term

survival in patients with diffuse peritoneal ovarian carcinomatosis. They achieved

high adequate primary and secondary surgical cytoreduction rates with acceptable

morbidity and mortality.

Verwaal et al (2008) noted that the treatment of PC is based on CRS followed by

HIPEC and combined with adjuvant chemotherapy. In 2003, a randomized trial

was finished comparing systemic chemotherapy alone with CRS followed

by HIPEC and systemic chemotherapy. This trial showed a positive result favoring

the studied treatment; and has now been updated to a minimal follow-up of 6

Page 10: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 10 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

years to show long-term results. For all patients still alive, the follow-up was

updated until 2007. In the original study, 4 patients were excluded -- 2 because of

no eligible histology/pathology and 2 because of major protocol violations. After

randomization, 4 patients in the HIPEC arm and 6 in the control arm were not

treated using the intended therapy, 1 patient because of withdrawal, 1 because of

a life-threatening other malignant disease and the others because of progressive

disease before initiation of the treatment. During the follow-up, 1 patient was

crossed-over from the control arm and underwent CRS and HIPEC for recurrent

disease, after the assigned treatment was completed. The data from these

patients were censored at the moment of the cross-over. Progression-free and

disease-specific survival were analyzed using the Kaplan Meyer test and

compared using the log rank method. The long-term results were studied in more

detail to evaluate efficacy and toxicity. At the time of this update, the median

follow-up was almost 8 years (range of 72 to 115 months). In the standard arm, 4

patients were still alive, 2 with and 2 without disease; in the HIPEC arm, 5 patients

were still alive, 2 with and 3 without disease. The median PFS was 7.7 months in

the control arm and 12.6 months in the HIPEC arm (p = 0.020). The median

disease-specific survival was 12.6 months in the control arm and 22.2 months in

the HIPEC arm (p = 0.028). The 5-year survival was 45 % for those patients in

whom a R1 resection was achieved. The authors concluded that with 90 % of all

events having taken place up to this time, this randomized trial shows that CRS

followed by HIPEC does significantly add survival time to patients affected by PC

of colorectal origin. For a selected group, there is a possibility of long-term

survival.

Elias and colleagues (2009) compared the long-term survival of patients with

isolated and resectable PC in comparable groups of patients treated with systemic

chemotherapy containing oxaliplatin or irinotecan or by CRS plus HIPEC. All

patients with gross PC from colorectal adenocarcinoma who had undergone CRS

plus HIPEC were evaluated. The standard group was constituted by selecting

patients with colorectal PC treated with palliative chemotherapy during the same

period, but who had not benefited from HIPEC because the technique was

unavailable in the center at that time. A total of 48 patients were retrospectively

included in the standard group and were compared with 48 patients who had

undergone HIPEC and were evaluated prospectively. All characteristics were

comparable except age and tumor differentiation. There was no difference in

systemic chemotherapy, with a mean of 2.3 lines per patient. Median follow-up

was 95.7 months in the standard group versus 63 months in the HIPEC group.

Two-year and 5-year overall survival rates were 81 % and 51 % for the HIPEC

group, respectively, and 65 % and 13 % for the standard group, respectively.

Median survival was 23.9 months in the standard group versus 62.7 months in the

HIPEC group (p < 0.05, log-rank test). The authors concluded that patients with

isolated, resectable PC achieve a median survival of 24 months with modern

chemotherapies, but only CRS plus HIPEC is able to prolong median survival to

roughly 63 months, with a 5-year survival rate of 51 %.

In a phase II clinical study, Lim et al (2009) evaluated the toxicities and treatment

response of intra-operative HIPEC in patients with advanced epithelial ovarian

cancer. Intra-operative HIPEC (cisplatin 75 mg/m(2), 41.5 degrees C, 90 mins)

was performed in 30 patients with residual tumor of less than 1 cm after CRS

between January 2007 and February 2008. All the patients received adjuvant

Page 11: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 11 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

chemotherapy with combination platinum and taxane. Adverse events and

responses to primary treatment were evaluated and scored as follows: grade I,

observation; grade II, medical treatment; grade III, intervention; and grade IV, re-

operation or admission to the intensive care unit. No deaths or grade IV

morbidities were observed. A total of 107 adverse events were identified in 30

patients (grade I, 40; grade II, 46; grade III, 21). The most common adverse

events affected the hematological system (n = 26), followed by the gastro-

intestinal system (n = 23). Most adverse events were anemias requiring

transfusion and nausea/vomiting requiring medication. Twenty-eight patients (93

%) experienced complete remission, and 2 patients (7 %) had progressive

disease. The authors concluded that HIPEC after extensive CRS for ovarian

cancer is a procedure with acceptable morbidity that patients can tolerate. They

stated that more follow-up is needed to determine the effect of HIPEC on survival.

In a review on ovarian cancer, Hennessy and colleagues (2009) stated that early

data suggested that HIPEC is promising, but this treatment is still highly

investigational. Furthermore, Ferron et al (2009) noted that scientific evidence on

the use of HIPEC for ovarian cancer remains poor. Much more research is still

needed to elucidate unanswered questions. Before this technique can be routinely

used, some controversial aspects have to be defined: (i) which drug is the best to

deliver and at what temperature, (ii) is it necessary to use mono- or poly-

chemotherapy regimens, (iii) which is the time-point for HIPEC in the natural

history of ovarian cancer: at front-line therapy, at interval debulking following initial

neo-adjuvant chemotherapy, at consolidation following front-line therapy, or at the

time of recurrence. Chua and colleagues (2009) stated that a randomized trial is

needed to establish the role of HIPEC in ovarian cancer.

There is some evidence that cytoreductive surgery with intra-operative

chemotherapy and hyperthermia may be of help in some individuals with

peritoneal mesothelioma, which is a rare disease, but increasing in frequency.

The incidence is about 1 per 1,000,000 and approximately 1/5 to 1/3 of all

mesotheliomas are peritoneal. Because of its unusual nature, the disease has not

been clearly defined either in terms of its natural history, diagnosis, or

management.

Sebbag et al (2000) discussed results of treatment of 33 patients (10 women and

23 men) with peritoneal mesothelioma. Patients were treated by a uniform strategy

involving CRS with peritonectomy procedures and peri-operative intraperitoneal

chemotherapy (cisplatin, doxorubicin). Median survival was 31.0 months; overall

projected survival at 3 years was 56 %. The most significant positive predictive

factors of survival were: female sex (p = 0.003), low prior surgical score (p =

0.002), completeness of cytoreduction (p = 0.0002) and second

-look surgery (p = 0.019). The morbidity rate for this combined treatment was 33

% and the peri-operative mortality rate was 3 %. These investigators concluded

that although peritoneal mesothelioma is rare, progress in its management has

occurred. Survival has been extended and selection factors by which patients

may be allocated to aggressive management strategies have been defined.

Sugarbaker et al (2002) reviewed a single institution's experience with 51 patients

prospectively treated over the past decade with increasingly aggressive

local/regional protocols. Peritoneal mesothelioma patients generally present with

Page 12: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 12 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

2 types of symptoms and signs: (i) those with abdominal pain, usually localized

and related to a dominant tumor mass with little or no ascites, and (ii) those

without abdominal pain, but with ascites and abdominal distention. Pathologically,

a positive immunostain for calretinin has markedly increased the accuracy of

diagnosis. Prognosis as determined by clinical presentation, the completeness of

cytoreduction, and gender (females survive longer than males) appears to be

improved by the use of intraperitoneal chemotherapy. Over the past decade, the

management of these patients has evolved similarly to ovarian cancer treatment

and now involves CRS, heated intraoperative intraperitoneal chemotherapy (HIIC)

with cisplatin and doxorubicin, and early post-operative intraperitoneal paclitaxel.

These perioperative treatments are followed by adjuvant intraperitoneal paclitaxel

and second-look cytoreduction. Prolonged DFS and reduced adverse symptoms

with the current management strategy are documented by a high complete

response rate as assessed by a negative second-look. This multi-modality

treatment approach with cytoreductive surgery and intraperitoneal chemotherapy

has resulted in a median survival of 50 to 60 months. Peritoneal mesothelioma is

an orphan disease that is treatable with expectations for "potential" cure in a small

number of patients if diagnosed and treated early with definitive local/regional

treatments. A prolonged high quality of life is possible in the majority of patients.

Sethna et al (2003) described their experience with 5 cases (4 females and 1

male) of cystic peritoneal mesothelioma. All of these patients were treated

with CRS with peritonectomy procedures and HIIC. The authors concluded that

cystic peritoneal mesothelioma should no longer be referred to as "benign" cystic

mesothelioma. An aggressive approach with complete disease eradication is the

correct goal of treatment. From the authors' experience, CRS to remove all visible

tumor and intraperitoneal chemotherapy to control microscopic residual disease

will help patients with peritoneal cystic mesothelioma to remain symptom- and

disease-free over an extended time period with a single surgical intervention.

Disease eradication may prevent the transition to an aggressive and fatal disease

process.

Diffuse malignant peritoneal mesothelioma (DMPM) is a subset of peritoneal

mesothelioma with a poor clinical outcome. Deraco et al (2006) performed a

prognostic analysis in a cohort of DMPM patients treated homogeneously by CRS

and intraperitoneal hyperthermic perfusion (IPHP). A total of 49 DMPM patients

who underwent 52 consecutive procedures were enrolled onto the study.

Cytoreductive surgery was performed according to the peritonectomy technique,

and the IPHP was performed with cisplatin plus doxorubicin or cisplatin plus

mitomycin C. These investigators evaluated the correlation of the

clinicopathologic variables (previous surgical score, age, sex, performance status,

previous systemic chemotherapy, carcinomatosis extension, completeness of

cytoreduction, IPHP drug schedule, mitotic count [MC], nuclear grade, and

biological markers [epidermal growth factor receptor, p16, matrix

metalloproteinase 2 and matrix metalloproteinase 9]) with overall and progression-

free survival. The mean age was 52 years (range of 22 to 74 years). The mean

follow-up was 20.3 months (range of 1 to 89 months). Regarding the biological

markers, the rates of immunoreactivity of epidermal growth factor receptor, p16,

matrix metalloproteinase 2, and matrix metalloproteinase 9 were 94 %, 60 %, 100

%, and 85 %, respectively. The strongest factors influencing OS were

completeness of cytoreduction and MC, whereas those for PFS were performance

Page 13: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 13 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

status and MC. No biological markers were shown to be of prognostic value. The

authors concluded that completeness of cytoreduction, performance status, and

MC seem to be the best determinants of outcome. These data warrant

confirmation by a further prospective formal trial. No biological markers presented

a significant correlation with the outcome. The over-expression of epidermal

growth factor receptor, matrix metalloproteinase 2, and matrix metalloproteinase 9

and absent or reduced expression of p16 might be related to the underlining tumor

kinetics of DMPM and warrant further investigation with other methods.

Kusamura et al (2006) analyzed morbidity and mortality of CRS and IPHP in the

treatment of peritoneal surface malignancies. A total of 205 patients (50 with

peritoneal mesothelioma, 49 with pseudomyxoma peritonei, 41 with ovarian

cancer, 32 with abdominal sarcomatosis, 13 with colon cancer, 12 with gastric

cancer, and 8 with carcinomatosis from other origins) underwent 209 consecutive

procedures. Four patients underwent the intervention twice because of disease

relapse. There were 70 men and 135 women. Mean age was 52 years (range

of 22 to 76 years). Cytoreductive surgery was performed by using peritonectomy

procedures; IPHP through the closed abdomen technique was conducted with a

pre-heated (42.5 degrees C) perfusate containing cisplatin + mitomycin C or

cisplatin + doxorubicin. Major morbidity rate was 12 %. The most significant

complications were 23 anastomotic leaks or bowel perforations, 4 abdominal

bleeds, and 4 sepses. Operative mortality rate was 0.9 %. On logistic regression

model multi-variate analysis, extent of cytoreduction (odds ratio [OR], 2.88; 95 %

confidence interval [CI]: 1.29 to 6.40) and dose of cisplatin for IPHP greater than

or equal to 240 mg (OR, 3.13; 95 % CI: 1.24 to 7.90) were independent risk factors

for major morbidity. Ten patients presented with grade 3 to 4 toxicity. The authors

concluded that CRS + IPHP presented acceptable morbidity, toxicity, and mortality

rates, all of which support prospective phase III clinical trials.

According to the National Cancer Institute's guideline on the treatment

of malignant mesothelioma (2009), "[i]ntrapleural or intraperitoneal administration

of chemotherapeutic agents (e.g., cisplatin, mitomycin, and cytarabine) has been

reported to produce transient reduction in the size of tumor masses and temporary

control of effusions in small clinical studies. Additional studies are needed to

define the role of intracavitary therapy". Furthermore, although the

NCCN guidelines on malignant pleural mesothelioma (2011) recommend cisplatin

as a 1st-line drug for inoperable mesothelioma, there is no recommendation for

either hyperthermic or intrapleural administration.

Baratti et al (2010) noted that unlike novel molecular-targeted therapies for

metastatic gastrointestinal stromal tumors (GIST), conventional treatments for

peritoneal sarcomatosis (PS) are mostly ineffective. As with carcinomatosis of

epithelial origin, a rationale base supports an aggressive loco-regional treatment of

PS, but the use of CRS and HIPEC in this setting is still controversial. These

researchers assessed the outcome of clinically and pathologically homogeneous

subsets of patients with PS uniformly treated by CRS and HIPEC. A prospective

database of 37 patients who underwent CRS and close-abdomen HIPEC with

cisplatin and doxorubicin or mitomycin-C was reviewed. PS originated from GIST

(pre-imatinib era) in 8 patients, uterine leiomyosarcoma (ULS) in 11,

retroperitoneal liposarcoma (RPLP) in 13, and other sarcoma in 5. Cytoreductive

surgery was macroscopically complete in 28 patients (75.7 %). Operative

Page 14: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 14 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

mortality was 3.7 % and morbidity 21.6 %. After median follow-up of 104 (range of

1 to 131) months, peritoneal disease progression occurred in 16 patients, distant

metastases in 5, and both in 13. For all patients, median OS was 26.2 months; 7

patients were alive at 46 to 130 months (ULS, n = 4; RPLP, n = 2; GIST, n = 1).

Retroperitoneal liposarcoma had the best OS (median of 34 months) but 100 %

peritoneal relapse; GIST had dismal overall, local-regional-free and distant-free

survival; ULS had the higher proportion of long survivors and best local-regional-

free survival. The authors concluded that overall, results of CRS and HIPEC did

not compare favorably to those of conventional therapy. In a subgroup analysis,

the combined approach did not change GIST and RPLS natural history. The

interesting results with ULS may warrant further investigations.

Esquivel et al (2011) noted that CRS and HIPEC are being widely used in the

treatment of patients with peritoneal surface malignancies. The open procedure

has been associated with high grade III and IV morbidity and prolonged

hospitalization. In this study, these researchers evaluated laparoscopic CRS and

HIPEC in patients with limited peritoneal surface malignancies. Patients with

peritoneal surface malignancies and no gross evidence of carcinomatosis on the

computed tomographic scan were enrolled to undergo laparoscopic CRS and

HIPEC. They aimed to assess the feasibility, safety, and outcome of this

procedure. Post-operative complications were reported according to the National

Cancer Institute Common Toxicity Criteria. From October 2008 to January 2010, a

total of 14 patients were enrolled into the protocol. Of the 14 patients, 1 patient

was found with extensive carcinomatosis at the time of laparoscopy and had no

surgical procedure; 13 patients had a complete cytoreduction and HIPEC, 10 (77

%) laparoscopically and 3 (23 %) were converted to an open procedure. There

was 1 grade 3 morbidity (10 %) and 1 patient (10 %) in the laparoscopy group

experienced a grade 4 complication, needing a re-operation for an internal hernia.

Mean length of hospital stay was 6 days for those completed laparoscopically, 8

days for those converted to an open procedure and 8 days for a matched cohort of

patients with an upfront open procedure. The authors concluded that this initial

investigative stage demonstrated the feasibility and safety of CRS and HIPEC via

the laparoscopic route in selected patients with low-tumor volume and no small

bowel involvement mainly from appendiceal malignancies. They stated that longer

follow-up and additional studies are needed to evaluate its long-term

effectiveness.

In a systematic review, Lammers et al (2011) evaluate the effectiveness of

chemohyperthermia (C-HT) as a treatment for non-muscle-invasive bladder cancer

(NMIBC). The review process followed the Preferred Reporting Items for

Systematic Reviews and Meta-analyses (PRISMA) guidelines. An electronic

search of the Medline, Embase, Cochrane Library, CancerLit, and

ClinicalTrials.gov databases was undertaken. Relevant conference abstracts and

urology journals were also searched manually. Two reviewers independently

reviewed candidate studies for eligibility and abstracted data from studies that met

inclusion criteria. The primary end-point was time to recurrence. Secondary end-

points included time to progression, bladder preservation rate, and adverse event

(AE) rate. A total of 22 studies met inclusion criteria and underwent data

extraction. When possible, data were combined using random effects meta-

analytic techniques. Recurrence was seen 59 % less after C-HT than after

mitomycin C (MMC) alone. Due to short follow-up, no conclusions can be drawn

Page 15: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 15 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

about time to recurrence and progression. The overall bladder preservation rate

after C-HT was 87.6 %. This rate appeared higher than after MMC alone, but valid

comparison studies were lacking. Adverse events were higher with C-HT than

with MMC alone, but this difference was not statistically significant. The authors

concluded that published data suggested a 59 % relative reduction in NMIBC

recurrence when C-HT is compared with MMC alone. Chemohyperthermia also

appears to improve bladder preservation rate. However, due to a limited number

of randomized trials and to heterogeneity in study design, definitive conclusions

can not be drawn. In the future, C-HT may become standard therapy for high-risk

patients with recurrent tumors, for patients who are unsuitable for radical

cystectomy, and in cases for which bacillus Calmette-Guérin treatment is

contraindicated.

Hurwitz et al (2011) presented long-term results from a phase II study that

evaluated the effectiveness of transrectal ultrasound hyperthermia plus radiation

with or without androgen suppression for the treatment of locally advanced

prostate cancer. Patients with clinical T2b-T3bN0M0 disease (according to 1992

American Joint Committee on Cancer [AJCC] criteria) received radiation plus 2

transrectal ultrasound hyperthermia treatments. After the first 4 patients, 6 months

of androgen suppression were allowed. The study was designed to assess

absolute improvement in the 2-year disease-free survival rate compared with the

short-term androgen suppression arm in Radiation Therapy Oncology Group

(RTOG) study 92-02. A total of 37 patients received a total of 72 hyperthermia

treatments. The mean cumulative equivalent minutes (CEM) Temp = 43°C was

8.4 minutes. According to the 1992 AJCC classification, there were 19 patients

with T2b tumors, 8 patients with T2c tumors, 5 patients with T3a tumors, and 5

patients with T3b tumors. The median Gleason score was 7 (range of 6 to 9), and

the median prostate-specific antigen (PSA) level was 13.3 ng/ml (range of 2 to 65

ng/ml). Thirty-three patients received androgen suppression. At a median follow-

up of 70 months (range of 18 to 110 months), the 7-year OS rate was 94 %, and

61 % of patients remained failure free (according to the American Society for

Therapeutic Radiology and Oncology definition for failure free survival). The

absolute rate of DFS at 2 years, which was the primary study end-point, improved

significantly (84 %) compared with a rate of 64 % for similar patients on the 4-

month androgen suppression arm of RTOG 92-02. When Phoenix criteria (PSA

nadir + 2 ng/ml) were used to define biochemical failure, 89 % of patients were

failure-free at 2 years. The authors concluded that hyperthermia combined with

radiation for the treatment of locally advanced prostate cancer appeared to be

promising. The current results indicated that further study of hyperthermia for the

treatment of prostate cancer with optimal radiation and systemic therapy is

warranted.

Suzuki et al (2000) noted that since clear cell carcinoma of the ovary does not

respond to conventional platinum-based chemotherapy, the prognosis of recurrent

tumors is especially poor. In a 51-year old female who underwent surgery for

clear cell carcinoma of the ovary, a solitary metastatic carcinoma developed in the

pelvic cavity 7 months after the initial surgery. The patient underwent a whole

pelvic irradiation at a total dose of 65 Gy combined with hyperthermia. Complete

remission was achieved 46 months after treatment. A study using gynecologic

carcinoma cell lines showed that the mean 50 % growth inhibitory dose of

radiation was 1.2 +/- 0.4 Gy in several clear cell carcinoma cell lines. The value

Page 16: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 16 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

did not significantly differ from those for serous carcinoma cell lines (2.3 +/- 1.2

Gy) and uterine cervical carcinoma cell lines (1.6 +/- 0.4 Gy). The authors stated

that currently, no anti-cancer agents are effective for clear cell carcinoma;

radiotherapy combined with hyperthermia may be effective for localized tumors.

Desmoplastic small round cell tumor (DSRCT) is a rare intra-abdominal

mesenchymal tissue neoplasm in young patients and spreads through the

abdominal cavity. Desmoplastic small round cell tumor usually presents with

diffuse abdominal metastatic disease similar in gross appearance to

carcinomatosis. To date, very aggressive treatment programs have yielded dismal

outcomes.

Msika et al (2010) stated that prognosis of DSRCT is poor despite a multi-modal

therapy including chemotherapy, radiotherapy, and surgical cytoreduction.

Hyperthermic intra-peritoneal chemotherapy is considered as an additional

strategy in the treatment of peritoneal carcinomatosis; for this reason, these

investigators planned to treat selected cases of children with DRSCT using

surgical cytoreduction and HIPEC. Peritoneal disease extension was evaluated

according to Gilly classification. Surgical cytoreduction was considered as

completeness of cytoreduction -- stage 0 of Gilly classification (no macroscopic

disease); HIPEC was performed according to the open technique. These

researchers described 3 cases: the 2 first cases were realized for palliative

conditions and the last one was operated on with curative intent. There was no

post-operative mortality. One patient was re-operated for a gallbladder

perforation. There was no other complication related to HIPEC procedure. The

authors concluded that surgical cytoreduction and HIPEC provide a local

alternative approach to systemic chemotherapy in the control of microscopic

peritoneal disease in DRSCT, with an acceptable morbidity, and may be

considered as a potential beneficial adjuvant waiting for a more specific targeted

therapy against the fusion protein.

In a review on “Desmoplastic small round cell tumor: Current management and

recent findings”, Dufrense et al (2012) states that “Overall, data supporting the use

of HIPEC in patients with DSRCT is limited and this technique is not

recommended for the management of patients with DSRCT outside clinical trials”.

National Comprehensive Cancer Network clinical practice guideline on “Non-small

cell lung cancer” (NCCN, 2013) does not mention the use of hyperthermia as a

therapeutic option.

Mi and colleagues (2013) stated that adjuvant intraoperative hyperthermic

intraperitoneal chemotherapy (IHIC) is a therapy that combines thermotherapy and

intraperitoneal chemotherapy. It is theoretically powerful for patients with

advanced gastric cancer (AGC), but is there no evident advantage in clinical

practice. These researchers performed a meta-analysis to evaluate the safety and

effectiveness of adjuvant IHIC inpatients with resectable locally AGC, and

provided the reference for clinical practice and study. These investigators

searched the Cochrane Library, PubMed, Embase, Web of Science and Chinese

databases (Chinese BioMedical Literature Database (CBM), China National

Knowledge Infrastructure (CNKI) and Wanfang) electronically and also retrieved

papers from other sources (tracing related references and communication with

other authors). All relevant randomized controlled trials (RCTs) were collected to

Page 17: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 17 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

compare surgery combined with IHIC to surgery without IHIC for AGC. There were

no language restrictions. After independent quality assessment and data

extraction by 2 reviewers, meta-analysis was conducted by RevMan 5.1 software.

A total of 16 RCTs involving 1,906 patients were included. Compared with surgery

alone, combination therapy (surgery plus IHIC) was associated with a

signiï¬Â cant improvement in survival rate at 1 year (hazard ratio (HR) = 2.99; 95

% CI: 2.21 to 4.05; p < 0.00001), 2 years (HR = 2.43; 95 % CI: 1.81 to 3.26; p <

0.00001), 3 years (HR = 2.63; 95 % CI: 2.17 to 3.20; p < 0.00001), 5 years (HR =

2.49; 95 % CI: 1.97 to 3.14; p < 0.00001), and 9 years (HR = 2.14; 95 % CI: 1.38 to

3.32; p = 0.0007). Compared with surgery alone, combination therapy was

associated with a signiï¬Â cant reduction in recurrence rate at 2 years (RR = 0.42;

95 % CI: 0.29 to 0.61; p < 0.00001), 3 years (RR = 0.35; 95 % CI: 0.24 to 0.51; p <

0.00001) and 5 years (RR = 0.47; 95 % CI: 0.39 to 0.56; p < 0.00001).

Intraoperative hyperthermic intraperitoneal chemotherapy was not found to be

associated with higher risks of anastomotic leakage, ileus, bowel perforation,

myelosuppression, GI reaction and hypohepatia, but it increased the incidence of

abdominal pain (RR = 21.46; 95 % CI: 5.24 to 87.78; p < 0.00001). The authors

concluded that compared with surgery alone, surgery combined with IHIC can

improve survival rate and reduce the recurrence rate, with acceptable safety.

However, they stated that safety outcomes should be further evaluated by larger

samples and high quality studies. Furthermore, these investigators stated that

hyperthermia for intraperitoneal chemotherapy needs more clinical research.

Furthermore, the NCCN clinical practice guideline on “Gastric cancer” (Version

2.2013) does not mention the use of hyperthermic intraperitoneal chemotherapy

as a therapeutic option.

National Comprehensive Cancer Network's clinical practice guideline on

“Pancreatic adenocarcinoma” (Version 1.2014) does not mention the use

of hyperthermic intraperitoneal chemotherapy as a therapeutic option.

Tejani et al (2014) stated that treatment of advanced appendiceal adenocarcinoma

at NCCN’s member institutions commonly incorporates agents used for colorectal

cancer. Guidelines from the NCCN stated that small bowel and appendiceal

carcinoma may be treated with systemic chemotherapy according to the NCCN

guidelines for colon cancer (Version 2.2015), which states that "the panel considers

the treatment of disseminated carcinomatosis with cytoreductive surgery and

HIPEC to be investigational and does not endorse this therapy outside of a

clinical trial".

Signet ring cell carcinoma (SRCC) is a diffuse type of adenocarcinoma found most

often in the glandular cells of the stomach. van Oudheusden et al (2015) noted that

SRCC patients have a poor oncologic outcome. These researchers examined if

the potential drawbacks of HIPEC outweigh the benefits in patients with peritoneally

metastasized SRCC. Patients with PC of colorectal origin referred to

2 tertiary centers between April 2005 and December 2013 were identified and

retrospectively analyzed. Data were compared between SRCC histology and

other differentiations. A total of 351 patients were referred for CRS + HIPEC

among which 20 (5.7 %) patients were identified with SRCC histology. CRS +

HIPEC was performed in 16 of these 20 (80 %) and 252 out of the 331 remaining

patients (76.1 %). A higher proportion of patients in the SRCC-group were

Page 18: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 18 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

diagnosed with N2 stage (62.5 % versus 36.1 %, p = 0.04). A macroscopic

complete resection was achieved in 87.5 % and 97.2 %, respectively (p = 0.04).

Median survival was 14.1 months compared to 35.1 months (p < 0.01).

Recurrence occurred in 68.8 % of the SRCC patients and in 43.7 % of the other

histology patients (p = 0.05). The authors concluded that patients with SRCC and

PC treated with CRS + HIPEC have a poor median survival only slightly reaching

over 1 year. In the presence of other relative contraindications, SRCC histology

should refrain a surgeon from performing CRS and HIPEC.

Furthermore, NCCN’s clinical practice guideline on “Gastric cancer” (Version

1.2015) does not mention HIPEC as a therapeutic option.

A recent review by Elias et al (2014a) stated that HIPEC for neuroendocrine

tumors (NETs) is in the investigational stage. Furthermore, Elias et al (2014b)

compared the results of complete cytoreductive surgery (CCRS) of peritoneal

metastases from NET with and without HIPEC and reported that they were not

able to determine whether HIPEC had a positive or negative impact on outcomes.

Guidelines from the National Cancer Institute on “Gastrointestinal Carcinoid

Tumors Treatment (PDQ®)” (2014) have no recommendation for HIPEC. Also,

NCCN’s guideline on NETs (Version 1.2015) has no recommendations for the use

of HIPEC for carcinoid tumors.

Tabrizian et al (2014) presented a series of patients with peritoneal hepatocellular

carcinoma (HCC) treated with CRS +/- HIPEC and evaluated their

clinicopathologic characteristics and outcomes. Between 07/2007 to 08/2012, 14

patients with limited disease to the peritoneum underwent CRS; 7 of these patients

received additional HIPEC treatment. Primary end-point was OS. Operative

treatment was directed for metachronous peritoneal disease in the majority (92.8

%) of patients. Mean intra-operative PCI was 9.9 (± 8.3) and complete

mascroscopic cyto-reduction (CCR 0-1) was achieved in all but 1 case. Overall

major morbidity rate (Clavien-Dindo III-IV) at 30 days was 7.1 %. One post-

operative death occurred in a patient with extensive tumor burden (PCI = 33,

CCR2). Median follow-up after initial surgery was 43.8 months and the median

time to metachronous peritoneal recurrence was 23 months. Three-year

recurrence rate after peritoneal resection was 100 %. Median survival of the

cohort CCR0-1 was 35.6 months. The authors concluded that treatment of

peritoneal HCC remains challenging and survival is poor. In well-selected

candidates, however, CRS +/- HIPEC may prolong survival compared to systemic

therapy alone in patients with peritoneal HCC.

However, NCCN’s clinical practice guideline on “Hepatobiliary cancers” (Version

1.2015) does not mention HIPEC as a therapeutic option.

A recently published review of small intestine neoplasms (Reynolds et al, 2014)

stated that the evidence for HIPEC in small bowel adenocarcinoma is anecdotal.

The review stated that "The role of more radical resections or metastasectomy for

small bowel adenocarcinoma is unclear but anecdotal reports indicate a possible

role for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy".

An UpToDate review on “Treatment of small bowel neoplasm” (Cusack and

Overman, 2014) stated that “Long-term survival has been reported after

Page 19: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 19 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

aggressive cytoreduction surgery and intraperitoneal hyperthermic chemotherapy

in a handful of highly selected patients with peritoneal carcinomatosis from a small

bowel adenocarcinoma. Experience with this approach, which is more commonly

applied to patients with pseudomyxoma peritonei or malignant peritoneal

mesothelioma, is very limited. Patients being considered for this approach should

be referred to a center with expertise in the management of peritoneal surface

malignancies". The authors recommended systemic chemotherapy for patients

with locally advanced unresectable or metastatic small bowel adenocarcinoma.

Also, guidelines from the NCCN stated that small bowel and appendiceal

carcinoma may be treated with systemic chemotherapy according to the NCCN

guidelines for colon cancer (Version 2.2015); which states that "the panel

considers the treatment of disseminated carcinomatosis with cytoreductive surgery

and HIPEC to be investigational and does not endorse this therapy outside of a

clinical trial".

An UpToDate review on “Clinical presentation and management of thymoma and

thymic carcinoma” (Bezjak et al, 2014) does not mention the use of hyperthermic

administration of intraperitoneal chemotherapy as a therapeutic option.

Furthermore, the NCCN’s clinical practice guideline on “Thymomas and thymic

carcinoma” (version 1.2014) does not mention the use of hyperthermic

administration of intraperitoneal chemotherapy as a therapeutic option.

CPT Codes / HCPCS Codes / ICD-9 Codes

Hyperthermia:

CPT codes covered if selection criteria are met:

77600 Hyperthermia, externally generated; superficial (i.e., heating to

a depth of 4 cm or less)

77610 Hyperthermia generated by interstitial probe(s); 5 or fewer

interstitial applicators

77615 more than 5 interstitial applicators

CPT codes not covered for indications listed in the CPB:

77605 Hyperthermia, externally generated; deep (i.e., heating to

depths greater than 4 cm)

77620 Hyperthermia generated by intracavitary probe(s)

Other HCPCS codes related to the CPB:

J8600 Melphalan, oral 2 mg

J9245 Injection, melphalan HCI, 50 mg

ICD-9 codes covered if selection criteria are met:

Page 20: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 20 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

172.0 - 172.9 Malignant melanoma of skin

174.0 - 175.9 Malignant neoplasm of breast

195.0 Malignant neoplasm of head, face, and neck

196.0 Secondary and unspecified malignant neoplasm of lymph

nodes of head, face, and neck

ICD-9 codes not covered for indications listed in the CPB (not all-

inclusive):

158.8 Malignant neoplasm of specified parts of peritoneum

160.2 - 160.5 Malignant neoplasm of maxillary, ethmoidal, frontal, and

sphenoidal sinuses

162.2 - 162.9 Malignant neoplasm of lung

171.0 - 171.9 Malignant neoplasm of connective and other soft tissue

[desmoplastic small round cell tumor]

183.0 Malignant neoplasm of ovary [clear cell carcinoma]

183.3 Malignant neoplasm of broad ligament of uterus (mesovarium;

parovarian region)

183.8 Malignant neoplasm of other specified sites of uterine adnexa

186.0 - 186.9 Malignant neoplasm of testis

197.6 Secondary malignant neoplasm of retroperitoneum and

peritoneum

198.6 Secondary malignant neoplasm of ovary

Cytoreductive surgery combined with hyperthermic intraperitoneal

chemotherapy:

CPT codes covered if selection criteria are met::

77605 Hyperthermia, externally generated; deep (i.e., heating to

depths greater than 4 cm)

77620 Hyperthermia generated by intracavitary probe(s)

96446 Chemotherapy administration into the peritoneal cavity via

indwelling port or catheter

CPT codes not covered for indications listed in the CPB:

96440 Chemotherapy administrations into pleural cavity, requiring and

including thoracentesis

ICD-9 codes covered if selection criteria are met:

Page 21: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 21 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

158.0 - 158.9 Malignant neoplasm of retroperitoneum and peritoneum

[peritoneal mesothelioma]

197.6 Secondary malignant neoplasm of retroperitoneum and

peritoneum [pseudomyxoma peritonei]

ICD-9 codes not covered for indications listed in the CPB (not all-

inclusive):

151.0 - 151.9 Malignant neoplasm of stomach

153.0 -153.9 Malignant neoplasm of colon

154.0 - 154.8 Malignant neoplasm of rectum, rectosigmoid junction, and

anus

157.0 - 157.9 Malignant neoplasm of pancreas

163.0 - 163.9 Malignant neoplasm of pleura [mesothelioma]

182.0-182.8 Malignant neoplasm of body of uterus [leiomyosarcoma]

188.0-188.9 Malignant neoplasm of bladder

Intraluminal hyperthermia:

No specific code

Transrectal ultrasound hyperthermia - no specific code:

ICD-9 codes not covered for indications listed in the CPB (not all-

inclusive):

185 Malignant neoplasm of prostate

The above policy is based on the following references:

1. Shidnia H, Hornback NB, Shen RN, et al. An overview of the role of

radiation therapy and hyperthermia in treatment of malignant melanoma.

Adv Exp Med Biol. 1990;267:531-545.

2. Green I. Hyperthermia alone or combined with chemotherapy for treatment

of cancer. AHCPR Health Technology Reports. 1991;(2):1-16.

3. Hafstrom L, Rudenstam CM, Blomquist E, et al. Regional hyperthermic

perfusion with melphalan after surgery for recurrent malignant melanoma. J

Clin Oncol. 1991;9(12):2091-2094.

4. Perez CA, Pajak T, Emami B, et al. Randomized phase III study comparing

irradiation and hyperthermia with irradiation alone in superficial measurable

tumors. Final report by the Radiation Therapy Oncology Group. Am J Clin

Oncol. 1991;14(2):133-141.

5. Seegenschmiedt MH, Sauer R, Fietkau R, et al. Interstitial thermal radiation

therapy: Five-year experience with head and neck tumors. Radiology.

1992;184(3):795-804.

Page 22: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 22 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

Kodama K, Doi O, Higashiyama M, et al. Long-term results of postoperative

intrathoracic chemo-thermotherapy for lung cancer with pleural

dissemination. Cancer. 1993;72(2):426-431.

Engin K, Leeper DB, Tupchong L, et al. Thermoradiation therapy for

superficial malignant tumors. Cancer. 1993;72(1):287-296.

Engin K, Tupchong L, Waterman FM, et al. Hyperthermia and radiation in

advanced malignant melanoma. Int J Radiat Oncol Biol Phys. 1993;25

(1):87-94.

Hoekstra HJ, Schraffordt Koops H, de Vries LG, et al. Toxicity of

hyperthermic isolated limb perfusion with cisplatin for recurrent melanoma

of the lower extremity after previous perfusion treatment. Cancer. 1993;72

(4):1224-1229.

Overgaard J, Gonzalez Gonzalez D, Hulshof MC, et al. Randomised trial of

hyperthermia as adjuvant to radiotherapy for recurrent or metastatic

malignant melanoma. European Society for Hyperthermic Oncology.

Lancet. 1995;345(8949):540-543.

Vernon CC, Hand JW, Field SB, et al. Radiotherapy with or without

hyperthermia in the treatment of superficial localized breast cancer: Results

from five randomized controlled trials. International Collaborative

Hyperthermia Group. Int J Radiat Oncol Biol Phys. 1996;35(4):731-744.

Emami B, Scott C, Perez CA, et al. Phase III study of interstitial

thermoradiotherapy compared with interstitial radiotherapy alone in the

treatment of recurrent or persistent human tumors. A prospectively

controlled randomized study by the Radiation Therapy Group. Int J Radiat

Oncol Biol Phys. 1996;34(5):1097-1104.

Urano M, Kuroda M, Nishimura Y. For the clinical application of

thermochemotherapy given at mild temperatures. Int J Hyperthermia.

1999;15(2):79-107.

Fraker DL. Hyperthermia regional perfusion for melanoma and sarcoma of

the limbs. Curr Probl Surg. 1999;36(110):841-907.

Sugarbaker PH, Chang D. Results of treatment of 385 patients with

peritoneal surface spread of appendiceal malignancy. Ann Surg Oncol.

1999;6(8):727-731.

van der Zee J, González González D, van Rhoon GC, et al. Comparison of

radiotherapy alone with radiotherapy plus hyperthermia in locally advanced

pelvic tumours: A prospective, randomised, multicentre trial. Dutch Deep

Hyperthermia Group. Lancet. 2000;355(9210):1119-1125.

Harima Y, Nagata K, Harima K, et al. A randomized clinical trial of radiation

therapy versus thermoradiotherapy in stage IIIB cervical carcinoma. Int J

Hyperthermia. 2001;17(2):97-105.

Reintgen D, Cruse CW, Atkins M. Cutaneous malignant melanoma. Clin

Dermatol. 2001;19(3):253-261.

Taylor ME. Breast cancer: Chest wall recurrences. Curr Treat Options

Oncol. 2002;3(2):175-177.

Wust P, Hildebrandt B, Sreenivasa G, et al. Hyperthermia in combined

treatment of cancer. Lancet Oncol. 2002;3(8):487-497.

van der Zee J. Heating the patient: A promising approach? Ann Oncol.

2002 Aug;13(8):1173-1184.

Page 23: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 23 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

California Technology Assessment Forum (CTAF). Isolated limb perfusion

for malignant melanoma of the extremity. Technology Assessment. San

Francisco, CA: CTAF; February 13, 2002.

National Comprehensive Cancer Network (NCCN), Gastric Cancer Panel.

Gastric adenocarcinoma. Clinical Practice Guidelines in Oncology --

v.1.2003. Rockledge, PA: NCCN; 2003.

Cancer Care Ontario. Adjuvant therapy for stage III colon cancer following

complete resection. Practice Guidelines #2-2. Toronto, ON: Cancer Care

Ontario; April 2000.

Cancer Care Ontario. Adjuvant therapy for stage II colon cancer following

complete resection. Practice Guidelines #2-1. Toronto, ON: Cancer Care

Ontario; April 2000.

Gurkan Zorlu C, Eylem Seker Ari P. Hyperthermia in gynecologic cancers.

Eur J Gynaecol Oncol. 2003;24(3-4):282-286.

Hehr T, Wust P, Bamberg M, Budach W. Current and potential role of

thermoradiotherapy for solid tumours. Onkologie. 2003;26(3):295-302.

Deraco M, Raspagliesi F, Kusamura S. Management of peritoneal surface

component of ovarian cancer. Surg Oncol Clin N Am. 2003;12(3):561-583.

Ryu KS, Kim JH, Ko HS, et al. Effects of intraperitoneal hyperthermic

chemotherapy in ovarian cancer. Gynecol Oncol. 2004;94(2):325-332.

Douwes F, BogoviC J, Douwes O, et al. Whole-body hyperthermia in

combination with platinum-containing drugs in patients with recurrent

ovarian cancer. Int J Clin Oncol. 2004;9(2):85-91.

Vasanthan A, Mitsumori M, Park JH, et al. Regional hyperthermia combined

with radiotherapy for uterine cervical cancers: A multi-institutional

prospective randomized trial of the international atomic energy agency. Int J

Radiat Oncol Biol Phys. 2005;61(1):145-153.

Bryant J, Clegg AJ, Sidhu MK, et al. Clinical effectiveness and costs of the

Sugarbaker procedure for the treatment of pseudomyxoma peritonei.

Health Technol Assess. 2004;8(7):1-66.

National Institute for Clinical Excellence (NICE). Complete cytoreduction

and heated intraoperative intraperitoneal chemotherapy (Sugarbaker

technique) for peritoneal carcinomatosis. Interventional Procedure

Guidance 116. London, UK: NICE; March 2005. Available at:

http://www.nice.org.uk/nicemedia/pdf/ip/IPG116guidance.pdf. Accessed

March 23, 2005.

National Institute for Clinical Excellence. Complete cytoreduction for

pseudomyxoma peritonei (Sugarbaker technique). Interventional Procedure

Guidance 56. London, UK: NICE; April 2004. Available at:

http://www.nice.org.uk/nicemedia/pdf/IPG056guidance.pdf. Accessed May

27, 2005.

Richel O, Zum Vörde Sive Vörding PJ, Rietbroek R, et al. Phase II study of

carboplatin and whole body hyperthermia (WBH) in recurrent and

metastatic cervical cancer. Gynecol Oncol. 2004;95(3):680-685.

Kouloulias V, Plataniotis G, Kouvaris J, et al. Chemoradiotherapy combined

with intracavitary hyperthermia for anal cancer: Feasibility and long-term

results from a phase II randomized trial. Am J Clin Oncol. 2005;28(1):91-99.

Jones EL, Oleson JR, Prosnitz LR, et al. Randomized trial of hyperthermia

and radiation for superficial tumors. J Clin Oncol. 2005;23(13):3079-3085.

Page 24: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 24 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

Stewart JH 4th, Shen P, Levine EA. Intraperitoneal hyperthermic

chemotherapy for peritoneal surface malignancy: Current status and future

directions. Ann Surg Oncol. 2005;12(10):765-777.

Westermann AM, Jones EL, Schem BC, et al. First results of triple-modality

treatment combining radiotherapy, chemotherapy, and hyperthermia for the

treatment of patients with stage IIB, III, and IVA cervical carcinoma. Cancer.

2005;104(4):763-770.

Issels RD, Schlemmer M, Lindner LH. The role of hyperthermia in

combined treatment in the management of soft tissue sarcoma. Curr Oncol

Rep. 2006;8(4):305-309.

Bergs JW, Franken NA, Haveman J, et al. Hyperthermia, cisplatin and

radiation trimodality treatment: A promising cancer treatment? A review

from preclinical studies to clinical application. Int J Hyperthermia. 2007;23

(4):329-341.

Mitsumori M, Zeng ZF, Oliynychenko P, et al. Regional hyperthermia

combined with radiotherapy for locally advanced non-small cell lung

cancers: A multi-institutional prospective randomized trial of the

International Atomic Energy Agency. Int J Clin Oncol. 2007;12(3):192-198.

Maluta S, Dall'Oglio S, Romano M, et al. Conformal radiotherapy plus local

hyperthermia in patients affected by locally advanced high risk prostate

cancer: Preliminary results of a prospective phase II study. Int J

Hyperthermia. 2007;23(5):451-456.

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(20):3737-3743.

Esquivel J, Sticca R, Sugarbaker P, et al; Society of Surgical Oncology

Annual Meeting. Cytoreductive surgery and hyperthermic intraperitoneal

chemotherapy in the management of peritoneal surface malignancies of

colonic origin: A consensus statement. Society of Surgical Oncology. Ann

Surg Oncol. 2007;14(1):128-133.

National Comprehensive Cancer Network (NCCN). Colon cancer. NCCN

Clinical Practice Guidelines in Oncology. v.2.2009. Fort Washington, PA:

NCCN; 2009.

National Comprehensive Cancer Network (NCCN). Breast cancer. NCCN

Clinical Practice Guidelines in Oncology. v.1.2009. Fort Washington, PA:

NCCN; 2009.

National Comprehensive Cancer Network (NCCN). Cervical cancer. NCCN

Clinical Practice Guidelines in Oncology. v.2.2009. Fort Washington, PA:

NCCN; 2009.

National Cancer Institute (NCI). Cervical cancer treatment (PDQ). Health

Professional Version. PDQ Cancer Information Summaries: Adult

Treatment. Bethesda, MD: NCI; updated May 16, 2008.

Franckena M, Stalpers LJ, Koper PC, et al. Long-term improvement in

treatment outcome after radiotherapy and hyperthermia in locoregionally

advanced cervix cancer: An update of the Dutch Deep Hyperthermia Trial.

Int J Radiat Oncol Biol Phys. 2008;70(4):1176-1182.

Minicozzi A, Borzellino G, Momo EN, et al. Treatment of the peritoneal

carcinomatosis by cytoreductive surgery and intraperitoneal hyperthermic

Page 25: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 25 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

chemotherapy (IHPC): Postoperative morbidity and mortality and short-term

follow-up. Ann Ital Chir. 2008;79(4):231-239.

Scaringi S, Kianmanesh R, Sabate JM, et al. Advanced gastric cancer with

or without peritoneal carcinomatosis treated with hyperthermic

intraperitoneal chemotherapy: A single western center experience. Eur J

Surg Oncol. 2008;34(11):1246-1252.

Spiliotis J, Tentes AA, Vaxevanidou A, et al. Cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy in the management of

peritoneal carcinomatosis. Preliminary results and cost from two centers in

Greece. J BUON. 2008;13(2):205-210.

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(2):315-325.

Verwaal VJ, Bruin S, Boot H, et al. 8-year follow-up of randomized trial:

Cytoreduction and hyperthermic intraperitoneal chemotherapy versus

systemic chemotherapy in patients with peritoneal carcinomatosis of

colorectal cancer. Ann Surg Oncol. 2008;15(9):2426-2432.

Elias D, Honoré C, Ciuchendéa R, et al. Peritoneal pseudomyxoma:

Results of a systematic policy of complete cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy. Br J Surg. 2008;95(9):1164-

1171.

Elias D, Lefevre JH, Chevalier J, et al. Complete cytoreductive surgery plus

intraperitoneal chemohyperthermia with oxaliplatin for peritoneal

carcinomatosis of colorectal origin. J Clin Oncol. 2009;27(5):681-685.

Lim MC, Kang S, Choi J, et al. Hyperthermic intraperitoneal chemotherapy

after extensive cytoreductive surgery in patients with primary advanced

epithelial ovarian cancer: Interim analysis of a phase II study. Ann Surg

Oncol. 2009;16(4):993-1000.

Hennessy BT, Coleman RL, Markman M. Ovarian cancer. Lancet.

2009;374(9698):1371-1382.

Ferron G, Martinez A, Mery E, et al. Importance of hyperthermic

intraperitoneal chemotherapy (HIPEC) in ovarian cancer. Bull Cancer.

2009;96(12):1243-1252.

Chua TC, Robertson G, Liauw W, et al. Intraoperative hyperthermic

intraperitoneal chemotherapy after cytoreductive surgery in ovarian cancer

peritoneal carcinomatosis: Systematic review of current results. J Cancer

Res Clin Oncol. 2009;135(12):1637-1645.

National Cancer Institute (NCI). Malignant mesothelioma treatment (PDQ).

Health Professional Information. Rockville, MD: NCI; January 9, 2009.

Available at:

http://www.cancer.gov/cancertopics/pdq/treatment/malignantmesothelioma/

healthprofessional/allpages. Accessed February 28, 2011.

Dovern E, de Hingh IH, Verwaal VJ, et al. Hyperthermic intraperitoneal

chemotherapy added to the treatment of ovarian cancer. A review of

achieved results and complications. Eur J Gynaecol Oncol. 2010;31(3):256-

261.

National Comprehensive Cancer Network (NCCN). Malignant pleural

mesothelioma. NCCN Clinical Practice Guidelines in Oncology. Version

I.2011. Fort Washington, PA: NCCN; 2011. Available at:

Page 26: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 26 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

65.

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

http://www.nccn.org/professionals/physician_gls/PDF/mpm.pdf. Accessed

February 28, 2011.

Deraco M, Baratti D, Laterza B, et al. Advanced cytoreduction as surgical

standard of care and hyperthermic intraperitoneal chemotherapy as

promising treatment in epithelial ovarian cancer. Eur J Surg Oncol. 2011;37

(1):4-9.

Sebbag G, Yan H, Shmookler BM, et al. Results of treatment of 33 patients

with peritoneal mesothelioma. Br J Surg. 2000;87(11):1587-1593.

Sugarbaker PH, Acherman YI, Gonzalez-Moreno S, et al. Diagnosis and

treatment of peritoneal mesothelioma: The Washington Cancer Institute

experience. Semin Oncol. 2002;29(1):51-61.

Sethna K, Mohamed F, Marchettini P, et al. Peritoneal cystic mesothelioma:

A case series. Tumori. 2003;89(1):31-35.

Deraco M, Nonaka D, Baratti D, et al. Prognostic analysis of

clinicopathologic factors in 49 patients with diffuse malignant peritoneal

mesothelioma treated with cytoreductive surgery and intraperitoneal

hyperthermic perfusion. Ann Surg Oncol. 2006;13(2):229-237.

Kusamura S, Younan R, Baratti D, et al. Cytoreductive surgery followed by

intraperitoneal hyperthermic perfusion: Analysis of morbidity and mortality

in 209 peritoneal surface malignancies treated with closed abdomen

technique. Cancer. 2006;106(5):1144-1153.

Baratti D, Pennacchioli E, Kusamura S, et al. Peritoneal sarcomatosis: Is

there a subset of patients who may benefit from cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy? Ann Surg Oncol. 2010;17

(12):3220-3228.

Chua TC, Liauw W, Zhao J, Morris DL. Upfront compared to delayed

cytoreductive surgery and perioperative intraperitoneal chemotherapy for

pseudomyxoma peritonei is associated with considerably lower

perioperative morbidity and recurrence rate. Ann Surg. 2011;253(4):769-

773.

Esquivel J, Averbach A, Chua TC. Laparoscopic cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy in patients with limited

peritoneal surface malignancies: Feasibility, morbidity and outcome in an

early experience. Ann Surg. 2011;253(4):764-768.

Lammers RJ, Witjes JA, Inman BA, et al. The role of a combined regimen

with intravesical chemotherapy and hyperthermia in the management of

non-muscle-invasive bladder cancer: A systematic review. Eur Urol.

2011;60(1):81-93.

Hurwitz MD, Hansen JL, Prokopios-Davos S, et al. Hyperthermia combined

with radiation for the treatment of locally advanced prostate cancer: Long-

term results from Dana-Farber Cancer Institute study 94-153. Cancer.

2011;117(3):510-516.

Suzuki M, Saga Y, Tsukagoshi S, et al. Recurrent ovarian clear cell

carcinoma: Complete remission after radiation in combination with

hyperthermia; a case study and in vitro study. Cancer Biother Radiopharm.

2000;15(6):625-628.

Msika S, Gruden E, Sarnacki S, et al. Cytoreductive surgery associated to

hyperthermic intraperitoneal chemoperfusion for desmoplastic round small

cell tumor with peritoneal carcinomatosis in young patients. J Pediatr Surg.

2010;45(8):1617-1621.

Page 27: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 27 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

Dufresne A, Cassier P, Couraud L, et al. Desmoplastic small round cell

tumor: Current management and recent findings. Sarcoma.

2012;2012:714986. Published online 2012 March 29.

National Comprehensive Cancer Network (NCCN). Melanoma. NCCN

Clinical Practice Guidelines in Oncology. v 2.2013.Fort Washington, PA:

NCCN; 2013.

National Comprehensive Cancer Network (NCCN). Non-small cell lung

cancer. NCCN Clinical Practrice Guidelines in Oncology. v.2.2013. Fort

Washington, PA: NCCN; 2013.

Mi DH, Li Z, Yang KH, et al. Surgery combined with intraoperative

hyperthermic intraperitoneal chemotherapy (IHIC) for gastric cancer: A

systematic review and meta-analysis of randomised controlled trials. Int J

Hyperthermia. 2013;29(2):156-167.

National Comprehensive Cancer Network (NCCN). Gastric cancer. NCCN

Clinical Practrice Guidelines in Oncology. v.2.2013. Fort Washington, PA:

NCCN; 2013.

National Comprehensive Cancer Network (NCCN). Pancreatic

adenocarcinoma. NCCN Clinical Practrice Guidelines in Oncology.

v.1.2014. Fort Washington, PA: NCCN; 2013.

Tabrizian P, Shrager B, Jibara G, et al. Cytoreductive surgery and

hyperthermic intraperitoneal chemotherapy for peritoneal carcinomatosis:

outcomes from a single tertiary institution. J Gastrointest Surg. 2014;18

(5):1024-1031.

Tejani MA, Ter Veer A, Milne D, et al. Systemic therapy for advanced

appendiceal adenocarcinoma: An analysis from the NCCN oncology

outcomes database for colorectal cancer. J Natl Compr Canc Netw.

2014;12(8):1123-1130.

Elias D, David A, Sourrouille I, et al. Neuroendocrine carcinomas: Optimal

surgery of peritoneal metastases and associated intra-abdominal

metastases. Surgery. 2014a;155(1):5-12.

Elias D, Goere D, Dumont F, et al. Role of hyperthermic intraoperative

peritoneal chemotherapy in the management of peritoneal metastases. Eur

J Cancer. 2014b;50(2):332-340.

Tabrizian P, Franssen B, Jibara G, et al. Cytoreductive surgery with or

without hyperthermic intraperitoneal chemotherapy in patients with

peritoneal hepatocellular carcinoma. J Surg Oncol. 2014;110(7):786-790.

Reynolds I, Healy P, McNamara DA. Malignant tumours of the small

intestine. Surgeon. 2014;12(5):263-270.

Cusack JC, Jr., Overman MJ. Treatment of small bowel neoplasms.

UpToDate Inc., Waltham, MA. Last reviewed December 2104.

Bezjak A, Meneshian A, Giaccone G. Clinical presentation and

management of thymoma and thymic carcinoma. UpToDate Inc., Waltham,

MA. Last reviewed December 2014.

National Comprehensive Cancer Network. Clinical practice guideline:

Thymomas and thymic carcinoma. Version 1.2014. NCN: Fort Washington,

PA.

National Cancer Institute. Gastrointestinal Carcinoid Tumors Treatment

(PDQ®). Last updated January 16, 2015. NCI: Bethesda, MD.

National Comprehensive Cancer Network. Clinical practice guidelines:

Hepatobiliary cancers. Version 1.2015. NCCN: Fort Washington, PA.

Page 28: Clinical Policy Bulletin: Hyperthermia in Cancer Therapy in Cancer Therapy Page 1 of 28 04/22/2015 Aetna …Published in: British Journal of Cancer · 1983Authors: N M BleehenAffiliation:

Hyperthermia in Cancer Therapy Page 28 of 28

http://qawww.aetna.com/cpb/medical/data/200_299/0278_draft.html 04/22/2015

95.

96.

97.

98.

National Comprehensive Cancer Network. Clinical practice guidelines:

Neuroendocrine tumors. Version 1.2015. NCCN: Fort Washington, PA.

National Comprehensive Cancer Network. Clinical practice guidelines:

Colon cancer. Version 2.2015. NCCN: Fort Washington, PA.

van Oudheusden TR, Braam HJ, Nienhuijs SW, et al. Poor outcome after

cytoreductive surgery and HIPEC for colorectal peritoneal carcinomatosis

with signet ring cell histology. J Surg Oncol. 2015;111(2):237-242.

National Comprehensive Cancer Network. Clinical practice guidelines:

Gastric cancer. Version 1.2015. NCCN: Fort Washington, PA.

Copyright Aetna Inc. All rights reserved. Clinical Policy Bulletins are developed by Aetna to assist in

administering plan benefits and constitute neither offers of coverage nor medical advice. This Clinical Policy

Bulletin contains only a partial, general description of plan or program benefits and does not constitute a

contract. Aetna does not provide health care services and, therefore, cannot guarantee any results or

outcomes. Participating providers are independent contractors in private practice and are neither

employees nor agents of Aetna or its affiliates. Treating providers are solely responsible for medical advice

and treatment of members. This Clinical Policy Bulletin may be updated and therefore is subject to change.

CPT only copyright 2008 American Medical Association. All Rights Reserved.