Extracranial oligometastatic renal cell carcinoma: current management

0 downloads 0 Views 2MB Size Report
The molecular fundamentals of oligometas- .... Extracranial oligometastatic renal cell carcinoma: current management & future directions REVIEW .... futuremedicine.com/doi/pdf/10.2217/fon.14.40 .... n th e ra p y trials in re nal ce ll ca rcin o m a . s tud y. (y e a r) s tu d y p e rio d. P ...... 15 Lussier YA, Xing HR, Salama JK et al.
Review

014

Extracranial oligometastatic renal cell carcinoma: current management and future directions

Jasmin Loh1, Ian D Davis2, Jarad M Martin1 & Shankar Siva*,3,4

1

4

2 1 7 /

uture

astatic nt man-

ture , UK

Abstract: The term ‘oligometastases’ was formulated to describe an intermediate state between widespread metastases and locally confined disease. The standard of care in metastatic renal cell carcinoma is systemic therapy; however, in patients with solitary or limited metastases, aggressive local therapies may potentially prolong survival. The literature suggests a survival benefit with surgical metastasectomy, with a reported 5-year survival as high as 45% in those who achieve complete resection. More recently, an expanding body of evidence supports the role of stereotactic ablative body radiation therapy for the treatment of oligometastatic renal cell carcinoma and early results demonstrate comparable local control rates with surgery. There is also increasing interest in the abscopal and immunologic effects of localized radiation. With the proliferation of newer targeted agents and immunomodulatory agents, current work is addressing the optimization of patient selection and avenues towards sequencing and combining the various treatment options. The concept of oligometastases, introduced by Hellman and Weichselbaum in 1995 describes an intermediate state of metastases in which the number and site of metastatic tumors are limited [1] . This clinical state includes two groups of patients: those with widespread and incurable, but mostly subclinical metastatic, disease; and those with truly limited metastatic disease and who may be potentially curable. The clinical implication of this hypothesis is that patients with overt metastases have distinct clinical and biological characteristics enabling them to be treated effectively with local ablative measures. The absolute number of metastatic lesions that defines the oligometastatic state is controversial. The precise definition has varied among the different studies. In general, oligometastases is defined as one to five metastases, involving either single or multiple organs, and are potentially amenable to local therapy with the aim of achieving long-term survival. There have been further suggestions by researchers from the University of Chicago (IL, USA) to distinguish between the different clinical scenarios that may exist within the concept of oligometastatic disease [2] . They include patients with:

Keywords 

• immunotherapy • metastasectomy • oligometastases • renal cell carcinoma • stereotactic

ablative body radiation therapy • targeted therapy

●● De novo oligometastases or synchronous disease: those that initially present with limited m ­ etastases,

although they may or may not also have more widespread subclinical metastatic disease [2] ; ●● Induced oligometastases or oligoprogressive disease: those who initially present with more wide-

spread metastases and who respond to systemic therapy, resulting in limited, but persistent, metastases [2] or differential limited progression in the face of good systemic control of other metastatic disease [3] ; Department of Radiation Oncology, Calvary Mater Hospital, Edith Street, Waratah NSW 2298, Australia Monash University Eastern Health Clinical School, Level 2, 5 Arnold Street, Box Hill, Melbourne, VIC 3128, Australia 3 Department of Radiation Oncology, Peter MacCallum Cancer Centre, Lansdowne Street, East Melbourne VIC 3002, Australia 4 Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, Melbourne, VIC, Australia *Author for correspondence: Tel.: +61 3 9656 1111; Fax: +61 3 9656 1424; [email protected] 1 2

10.2217/FON.14.40 © 2014 Future Medicine Ltd

Future Oncol. (2014) 10(5), 761–774

part of

ISSN 1479-6694

761

Review  Loh, Davis, Martin & Siva ●● Recurrent oligometastases or metachronous

disease: those who initially present with and are treated curatively for locoregional disease and subsequently develop further limited meta­ stases [4] . Niibe and Hayakawa coined the term oligorecurrence to describe an oligometastatic state where the primary site is c­ontrolled [5] . Approximately 25–30% of patients with newly diagnosed renal cell carcinoma (RCC) have metastatic disease at the initial presentation and approximately a third with clinically localized primary tumor at diagnosis will subsequently develop metastatic disease [6] . Historically, patients with metastatic RCC (mRCC) have a poor prognosis, with 5-year survival rates of ≤10% [6] . However, prolonged survival has been noted in patients with solitary or oligometastatic disease amenable to resection. With improved and more sensitive imaging methods, the oligo­metastatic state is increasingly being identified and, thus, the optimal management strategy in these patients warrants revisiting. RCC is often considered resistant to older conventional treatments including cytotoxic chemotherapy, conventionally fractionated radiation therapy and cytokine-based immunotherapy. This has led to surgery being considered as the only potential curative means in these patients. However, another local therapy is now available. Stereotactic ablative body radiation therapy (SABR) is able to deliver much higher biological doses of radiation therapy using high-precision techniques. Therefore, the conventional paradigm of surgical management of oligo-mRCC is now being challenged. The management of intracranial metastases has previously been published and, therefore, this review will focus exclusively on extracranial oligo-mRCC [7,8] . Biology of metastases Paget hypothesized in his ‘seed and soil hypothesis’ that metastases depend on interactions between tumor cell and target organ [9,10] . Certain tumors have a predilection to metastasize to particular organs and successful colonization of the target organ is owing to a complex interaction between the host, tumor microenvironment and genetic instability of the tumor cells. The discrete steps in the biological cascade of metastasis have been well described and have provided a framework to categorize the genes involved in each step of the metastatic process [11] . Adding to this complexity is the recently

762

Future Oncol. (2014) 10(5)

demonstrated intratumor heterogeneity by multiregion sequencing [12] . This implies that there are primary tumor subpopulations that only have a limited capability to metastasize, t­herefore, resulting in the oligometastatic state. The molecular fundamentals of oligometastases are still largely unknown. A panel of genes have been identified that were differentially expressed between patients with aggressive and nonaggressive RCC, but were not significantly different between aggressive primary and mRCC. These findings suggest that gene expression alterations that result in aggressive behavior and metastatic potential can be identified in the primary tumor [13] . Gene-expression profiling of resected pulmonary metastases from 18 patients with RCC using DNA microarrays found differential expression of 135 genes stratifying patients with fewer (≤8) versus multiple (≥16) metastases [14] . Furthermore, gene ontology enrichment analysis demonstrated upregulation of genes that positively regulate the cell cycle in those with multiple metastases, indicating increased growth potential in multiple metastases versus fewer metastases and hence implying a fundamental biological difference between these two clinical scenarios. More recently, miRNA expression has demonstrated the potential to identify patients most likely to remain oligometastatic after metastases-directed treatment [15] . Imaging Technological advances in imaging modalities have increased the frequency and likelihood of detecting oligometastatic disease. Routine use of cross-sectional imaging, such as computed tomography (CT) scanning, has improved the early detection of metastases. MRI has been shown to be highly sensitive and more specific compared with bone scintigraphy, with improved detection of bone metastases, as well as soft tissue disease [16] . Fluorine-18 fluorodeoxyglucose PET combined with CT has increased the detection of metastases, with the majority of cases reporting sensitivity close to a 100% for metastatic disease [17] . There is further work currently being carried out to assess utility of other molecular imaging, such as PET with novel radiolabelled isotopes. For example, cG250 is a monoclonal antibody targeting the carbonic anhydrase IX antigen, which is expressed in more than 90% of clear cell RCC and have preliminary data not only for diagnosis of primary and mRCC, but also as a therapeutic strategy [18] .

future science group

Extracranial oligometastatic renal cell carcinoma: current management & future directions  Together, these imaging modalities improve the accuracy of detecting an oligometastatic state, not only by detecting occult metastases, but also by detecting additional disease that would reclassify a patient into the broadly m­etastatic category. Metastasectomy There is a long history of surgery in the locally aggressive management of mRCC. Regarding extracranial lesions, the first pulmonary metastasectomy reported in 1945 [19] , with the patient dying 23 years after metastasectomy without evidence of recurrence . Resections for mRCC have been performed for solitary or limited metastatic disease to provide long-term disease control and a potential cure; recurrent disease, again with a view to long-term control or cure, or for palliation; symptomatic disease for local control and palliation of symptoms; and selected cases of limited residual disease following response to systemic therapy. The metastasectomy literature consists mainly of retrospective studies that are sometimes contradictory and suffer from selection biases; there has not been any published prospective randomized trial to date. Metastasectomy, in appropriately selected patients, has been shown to be feasible and with acceptable surgical morbidity. Pulmonary metastasectomy The lung is the most common site of metastases in mRCC. The 5-year overall survival rate after pulmonary metastasectomy ranges from 31 to 39% (Table 1) , and on subgroup analyses, complete resection has been consistently shown to be the strongest predictor of survival with reported survival rates up to 45% at 5 years [20–24] . Other prognostic factors associated with a favorable outcome include fewer number and/or smaller size of metastases, metachronous (versus synchronous) metastatic disease and negative hilar or mediastinal lymph node involvement at time of metastasectomy. Repeat thoracotomies for recurrent pulmonary metastases were not associated with an increase in morbidity or mortality and no survival difference was observed in these patients compared with those who were operated on only once, although patient selection would clearly be important [20,22,24] . Bone metastasectomy The second most common distant metastatic site of RCC is bone, with incidence rates of 20–25%

future science group

Review

at the time of initial presentation of metastatic disease [30] . The 5-year survival rates have been reported to be between 13 and 38% after resection of solitary osseous metastases (Table 1) , and patients with solitary metastases survive longer on average than those with multiple metastases [25–29] . Studies have shown conflicting results for the prognostic significance of the extent of surgery (radical vs marginal resection), localization of the osseous metastases (axial vs appendicular skeleton), timing of onset of metastases (synchronous vs metachronous) and presence of pathological fracture. The Fuhrman grade of the primary renal cell tumor was not found to be predictive of long-term survival in bone metastases [28,29] . Resection of other extracranial sites of metastasis Oligometastases to other extracranial sites, such as the thyroid, liver and pancreas, in RCC are uncommon. Published data on metastasectomy to these sites are, therefore, limited and there are no established guidelines for management. In a retrospective series of 45 patients, the 5-year survival rate following thyroid metastasectomy for mRCC was 51% [31] . Reported 5-year survival rates following liver metastasectomy for mRCC range from 26 to 62.2%, and prognostic factors that have been associated with improved survival include clear resection margins, metachronous rather than synchronous metastases, low grade of primary tumor, male sex and metastases diameter of ≤5 cm [32–34] . Several studies have reported 5-year survival rates of above 80% following pancreatic metastasectomy [35] , and a combined analysis of 15 studies reported a 5-year survival rate of 66% [36] . Data from the available literature support the role of resection of limited mRCC. However, these predominantly retrospective studies warrant validation in a prospective randomized manner. The prognostic significance of histological subtype on metastasectomy outcomes remains unclear. The predominant subtype is clear cell RCC, and the remaining subtypes including papillary, chromophobe, collecting duct and medullary cell types are grouped together as nonclear cell RCC. Most studies do not distinguish between clear and nonclear cell types. The limited retrospective data report contradictory findings. One study reported that the overall survival following pulmonary metastasectomy was not influenced by the histologic subtype (clear

www.futuremedicine.com

763

Review  Loh, Davis, Martin & Siva Table 1. Pulmonary and bone metastasectomy series for renal cell carcinoma. Study (year) 

Study period (years)

Total patients Nephrectomy Extrapulmonary†/ (complete performed extraosseous‡ resection), n metastases (patients, %) 

Pulmonary metastases Pfannschmidt 1985–1999 191 (149) et al. (2002)

Complete resection, negative LN involvement, number of metastases: ≤7, DFI: >23 months Complete resection, number of metastases: ≤6 Complete resection, negative LN involvement, number of metastases: 3 metastases) NA

NA

NA

All grade 1–2 toxicity

Toxicity

NA

72% at 1 year

49% at 1 year

NA

NA

None reported during the follow-up period

4% grade 3 toxicity (pain and fatigue). No grade 3–4 neurologic toxicity observed

Most common toxicity is grade 1 fatigue (10.5%). One case of grade 3 nausea and vomiting. No grade 4 toxicity

NA

No grade 2 or higher toxicity with RTOG/EORTC toxicity criteria

Two cases of grade 2 dermatitis, four developed fractures, one case of grade 4 erythema 32 months 96% had grade 1–2 toxicity. One death (patient treated for large metastatic lesion in lung close to pleura died 10 weeks after SABR after admission with electromechanical dissociation) NA 40% had grade ≥1 toxicity, majority had grade 3 toxicity. One death (fatal gastric hemorrhage 4 months after SABR for metastasis in pancreas adjacent to stomach and duodenum)

NA

NA (median 85% at DFS: 2-years 12.7 months)

Median survival

† Prospective trial. EORTC: European Organisation for Research and Treatment of Cancer; DFS: Disease-free survival; NA: Not available; RTOG: Radiation Therapy Oncology Group; SABR: Stereotactic ablative body radiation therapy.

48 (60)

Gerszten et al. (2005)

NA

Cervical (6), thoracic (26), lumbar (23)

Lung (63), kidney, adrenal, thoracic wall, abdominal glands, liver, pelvis, spleen Lung (117), kidney, renal bed, adrenal, pancreas, spleen, mediastinum, thoracic wall, bone, abdominal lymph nodes Orbits, head and neck, lung, mediastinum, sternum, clavicle, scapula, humerus, rib, spine, abdominal wall, primary kidney tumor Extracranial metastases (171) Primary kidney tumors (33) Spine

Osseous, abdominal lymph nodes, mediastinum/hilum, lung, kidney, adrenal, liver, soft tissue Pelvic bones, femur, spine, lymph nodes

Treated sites (treated lesions), n 

Nguyen et al. 2002–2007 48 (55) (2010)

57 (88)

1997–2005 92 (204)

NA

Balagamwala NA et al. (2012)

Gilson et al. (2006)

Teh et al. (2007)

Wersall et al. 1997–2003 58 (162) (2005)

Svedman et al.† (2006)

Zelefsky et al. 2004–2010 55 (105) (2012)

Ranck et al. (2013)

Patients (treated lesions), n 2006–2010 18 (39)

Study (year) Study period

Table 2. Stereotactic ablative body radiation therapy trials in renal cell carcinoma.

[48]

[47]

[46]

[45]

[44]

[43]

[42]

[41]

[40]

Ref.

Extracranial oligometastatic renal cell carcinoma: current management & future directions 

Review

767

Review  Loh, Davis, Martin & Siva Table 3. Targeted therapy trials in renal cell carcinoma. Targeted agents

Mechanism of action

Trials

Objective response rates (%) 

Median progression-free survival (months)

Median overall survival (months)

Ref.

Multikinase inhibitor targeting VEGFR, PDGFR, c-KIT Angiogenesis inhibitor targeting VEGFR, PDGFR and c-KIT mTOR inhibitor

Sunitinib vs IFN-α

31 vs 6

11.0 vs 5.0

26.4 vs 21.8 (ns)

[52]

Pazopanib vs placebo Pazopanib vs sunitinib

30 vs 3 31 vs 25

11.1 vs 2.8 8.4 vs 9.5

NA 28.4 v 29.3

[54]

Temsirolimus vs IFN-α vs both Bevacizumab plus IFN-α vs placebo plus IFN-α Bevacizumab plus IFN-α vs IFN-α Sorafenib vs IFN-α

9 vs 5 vs 8‡

3.8 vs 1.9 vs 3.7‡

10.9 vs 7.3 vs 8.4‡

[55]

31 vs 12

10.2 vs 5.4

23.3 vs 21.3 (ns)

[56]

26 vs 13

8.5 vs 5.2

18.3 vs 17.4

[57]

68 vs 39

5.7 vs 5.6

NA

[58]

Everolimus vs placebo Axitinib vs sorafenib

1 vs 0 (ns) 19 vs 9

4.0 vs 1.9 6.7 vs 4.7

14.8 vs 14.4 (ns) Data not mature

[60]

Sorafenib vs placebo

NA

5.5 vs 2.8

19.3 vs 15.9

[61]

First-line therapy Sunitinib† Pazopanib†   Temsirolimus† Bevacizumab†   Sorafenib§

Monoclonal antibody targeting VEGFR   Small molecule that inhibits multiple isoforms of intracellular serine/threonine kinase and other receptor tyrosine kinases

[53]

Second-line therapy Everolimus† Axitinib† Sorafenib†

mTOR inhibitor Second-generation VEGFR inhibitor Second-generation VEGFR inhibitor 

[59]

National Comprehensive Cancer Network category 1 recommendation. Statistically significant results between temsirolimus versus IFN-α only. National Comprehensive Cancer Network category 2A recommendation. NA: Not available; ns: Not statistically significant; PDGFR: PDGF receptor; VEGFR: VEGF receptor. † ‡

§

metastases. Most patients had undergone nephrectomy before targeted therapy in the trials that demonstrated the benefits of targeted agents (Table 3) . Cytoreductive nephrectomy, therefore, continues to be performed in appropriately selected patients. Two ongoing prospective randomized trials (the CARMENA trial [68] and the SURTIME trial [69]), investigating the role and timing of cytoreductive nephrectomy in patients receiving targeted therapy, respectively, are addressing this question. Similarly, there is paucity of data on the efficacy of metastasectomy in the targeted therapy era. A retrospective study by Johannsen et al. investigated discontinuation of targeted therapy in mRCC patients after a duration of therapy ranging from 4 to 26 months. The study focused on patients who achieved complete response with either targeted therapy alone or a combined approach of additional resection of residual metastases. These approaches were followed by recurrence in 66.7% of patients and re-exposure to targeted therapy was effective in 86.9% of cases. The median time without targeted therapies was 7 months [70] . A potential benefit of the

768

Future Oncol. (2014) 10(5)

combined approach is to enable patients a period of time without targeted therapies and thereby mitigate the duration of the sometimes debilitating associated side effects. The evidence for safety with combined modality management with surgery and systemic targeted agents is limited. This evidence is largely retrospective in nature. Increased risk of wound-related complications have been reported in a retrospective study of patients treated with various targeted agents prior to cytoreductive nephrectomy [71] . A prospective Phase II study that investigated the use of two to three cycles of sunitinib prior to cytoreductive nephrectomy reported surgical complication rates of 27% [72] . The majority (16%) had delayed wound healing and there was one death due to respiratory failure. An 18% postoperative complication rate was reported in a retrospective study of patients who underwent metastasectomy of various sites after receiving at least one cycle of targeted therapy [73] . The majority developed chylous ascites after retroperitoneal surgery. There were no bleeding or wound complications seen.

future science group

Extracranial oligometastatic renal cell carcinoma: current management & future directions  There is potential scope for integration of SABR with targeted therapy for synergistic effect. A Phase II trial investigating the feasibility of concurrent sunitinib and hypofractionated radiation therapy in 25 patients with oligometastases from various primary tumors reported local control rates of 75% at 18 months. The incidence of acute grade ≥3 toxicities was 28%, most commonly myelosuppression, bleeding and abnormal liver function test and there was one grade 5 hemorrhage. Caution will need to be used in these settings as many of the targeted therapies are radiosensitizers. A dose reduction is recommended for sunitinib when used concurrently with radiation therapy, and caution is advised in patients on any form of anticoagulation. Furthermore, the irradiation of large volumes particularly over sensitive structures such as small bowel with concurrent targeted agents is likely to increase the risk of toxicity [74] . Therefore, validation of clinical benefit of concurrent use compared with either SABR or targeted therapy alone in randomized trials is essential. Treatment of a lesion progressing on targeted therapy while other lesions remain controlled is reasonable in the light of the biological differences between metastases in the same patient. This approach also enables the patient to continue on that particular targeted agent and achieve the maximum clinical benefit before moving on to second-line therapy [3] . Conversely, if toxicities from systemic therapies are proving difficult to tolerate, consolidative radiation therapy to residual disease following response to t­argeted therapy may allow patients a drug holiday. Immunotherapy Defects in many aspects of the immune response are common in RCC [75] . Established tumors in the human host are capable of effective evasion of the tumor-directed immune response. In this setting, radiation therapy has been shown to elicit reactivation of the tumor-directed immune system [76] . The ‘abscopal effect’ is a bystander effect of radiation therapy observed as tumor regression remote from the site of irradiation. The abscopal effect is a rare event, but has been documented in a variety of tumors, including RCC [77] , and can occur with conventionally fractionated radiation therapy as well as SABR. A recent case report of an abscopal effect in a patient with melanoma treated with ipilimumab (an inhibitor of immunologic checkpoint on

future science group

Review

T cells) and radiation therapy has further fueled interest in this area [76] . Our own group has demonstrated abscopal effects secondary to SABR of a pulmonary metastasis in the context of nonsmall-cell lung cancer [78] . One series reported on four cases of RCC with an abscopal response out of 28 cases treated with SABR. In three out of the four cases, untreated metastatic disease completely regressed with no evidence of relapse at the time of reporting that ranged from 2 to 4 years post-therapy [79] . One of the proposed mechanisms for the abscopal effect of radiation therapy is the release of antigens and cytokines that can then inhibit tumor growth [80] . The primary driver of increased immune mediated cell death is an enhanced capacity to recognize and mount an adaptive immune response to the established tumor. Direct ionizing radiation elicits innate immune recognition of tumor, in the absence of a pathogen, through the liberation of cellular stress signals collectively termed, ‘danger signals’ [81,82] . In this context, radiation has been shown to profoundly augment T-cell priming and activation in human subjects, leading to elimination of cancer cells in a CD8 + T-cell-dependent manner [83] . Recent evidence suggests that ablative doses of radiation evoke a particularly strong immune response. Lee et al. demonstrated that T cells are required to elicit tumor responses after ablative radiation therapy [84] . Survival after ablative radiation therapy was decreased by more than 75% in mice deficient in CD8 + cells (the major killer T cells). Inhibition of tumor growth was more pronounced with a single dose of 20 Gy compared with a non­ablative dose of 5 Gy in four fractions delivered over 2 weeks. These results suggest that a radiation therapy-induced adaptive immune response can be a dose-dependent phenomenon and may result in additional tumor cell kill beyond direct radiation damage to DNA. Conventionally fractionated radiation therapy and adjuvant chemotherapy are speculated to be potentially immunosuppressive [84] . In the former, the fractionated low-dose radiation therapy may deplete radiosensitive T cells over time leading to suppression of the adaptive immune response. In the latter, chemotherapy abolished priming of the CD8 + T cells and forestalled cytotoxic T-cell proliferation, which may lead to increased rates of recurrence. By contrast, immunotherapy given in conjunction with radiation therapy amplified the radiation-mediated immune responses [84] .

www.futuremedicine.com

769

Review  Loh, Davis, Martin & Siva These findings have caused a resurgence of interest in exploring immunotherapy in RCC [85] . Recent work has demonstrated that immune tolerance of host tissues including cancer is actively regulated through various ‘immune checkpoint’ molecules, which act to inhibit immune responses including autoimmunity against cancer [85] . There are many such molecules and those of key clinical interest at present include CTLA-4 and PD-1. CTLA-4 moves rapidly to the cell surface on activation of T cells and inhibits T-cell activation. By contrast, PD-1 is expressed mainly on activated lymphocytes and, thus, is relevant at the time the T cell encounters the target it is meant to kill [86] . Activation of PD-1 at that time will protect the cancer from an effective immune response. Ligands for PD-1, such as PD-L1 and PD-L2, are expressed by many tissues including cancer cells. Antibodies against CTLA-4 are now approved for use (e.g., ipilimumab [Yervoy ®, Bristol-Myers Squibb, NJ, USA]) or under development (e.g., tremelimumab [Pfizer Inc./ Medimmune, NY, USA]). Antibodies against PD-1 (e.g., nivolumab [Bristol-Myers Squibb]) or PD-L1 (e.g., BMS-936559/MDX-1105 [BristolMyers Squibb] or MPDL3280A [Roche, NJ, USA]) are also under development. Nivolumab treatment led to objective responses in 27% of RCC patients with an acceptable adverse event profile in a Phase I study [87] . The complementary actions of CTLA-4 and PD-1 make it logical to combine therapies blocking these pathways or to combine them with other therapies. This approach has been shown to be effective in several clinical trials in a range of cancer types [87–90] . These approaches act on the host immune system and there is every reason to be confident that they will be effective in multiple tumor types. Several strategies are of relevance to RCC. For example, the trial that compares the anti-PD-1 antibody nivolumab with the mTOR inhibitor everolimus and has now completed accrual [91] . The anti-CTLA-4 antibody ipilimumab has demonstrated efficacy in RCC [92] . Interestingly, there may be other indirect interactions between therapies for RCC and the immune system. For example, sunitinib has been shown to decrease numbers of immune cells with immunosuppressive effects, prevent development of T-cell unresponsiveness, alter cytokine-expression profiles in favor of an anti-tumor effect, and improve survival of tumor-bearing mice [93] . This study used tumor models other than RCC, but is relevant due to

770

Future Oncol. (2014) 10(5)

the common use of sunitinib in RCC. At least one clinical trial is investigating various combinations of therapies including that of nivolumab plus sunitinib or pazopanib [94] . Other forms of cancer immunotherapy are also relevant in RCC. The cellular immunotherapy AGS-003 has shown promising results in a Phase II trial and a Phase III trial is under development. Seung et al. reported on a Phase I study of metastatic melanoma and RCC patients, in which patients were treated with SABR with one, two or three doses of 20 Gy per fraction followed by IL- 2 administration [95] . Response rates in nonirradiated lesions were higher than expected with combination treatment compared with IL-2 alone. These observations have led to intensified efforts to combine radiation therapy with an immune modifier to increase the effects of radiation therapy through immune responses, such as this trial investigating SABR and anti-CTLA-4 therapy [96] . Conclusion Although mRCC has in the past been associated with poor prognosis, some patients with solitary or limited metastatic disease can survive for long periods. Oligo-mRCC may, in some cases, be a biologically distinct entity with limited capacity to metastasize further. If that is the case then local aggressive therapy may lead to long-term disease control and potential cure. Currently surgery remains at the forefront of potentially curative locally ablative modalities for oligo-mRCC, but with increasing experience in SABR, the dogma that only surgery can afford a potential cure is being challenged. A key challenge continues to be the correct identification of patients who are most likely to benefit from aggressive local therapy. Although advances in imaging techniques have improved the detection of patients with a truly oligometastatic state, the incorporation of molecular classifiers of oligometastases can have a potential role in increasing the accuracy of the identification of such patients. High local control rates are seen with SABR and reported toxicity rates are low. However, in the absence of randomized data, the benefits of SABR or surgery have yet to be validated. Although randomized controlled trials provide the strongest evidence, due to the uncommon scenario and variety of presentations of the oligometastatic state, it can be difficult to conduct such studies in this patient population. Other trial designs that may be more realistic and reflective of the heterogeneity of RCC and its presentations

future science group

Extracranial oligometastatic renal cell carcinoma: current management & future directions  include N of 1 studies, tissue-based approaches, propensity matched analyses and adaptive design methods. The optimal strategies for combining the different modalities of treatment (surgery, SABR and systemic therapy) in mRCC have yet to be established and current trials are ongoing. Future perspective With increasing use and experience, and in the right context, SABR may be a viable alternative to surgery as local therapy in the management

Review

of oligo-mRCC, particularly as follow-up data matures and the high local control rates are maintained. However, the current experience with SABR is still limited and the duration of patient follow-up is shorter compared with surgery. It is imperative to ensure that careful validation of SABR is performed, including documentation of practice, outcomes, and quality measures; and that appropriate clinical trials are performed in order to provide high-level evidence of its effect before SABR is widely adopted. Further opportunities to

Executive summary Definition ●●

Oligometastases describes an intermediate state of cancer spread in which the number and site of metastatic tumors are limited. The clinical implication of this is that local therapy may be potentially curative.

●●

The clinical entity of oligometastatic renal cell carcinoma is increasingly being recognized, and a subset of patients can potentially achieve long-term survival with aggressive local therapy.

Metastasectomy ●●

Surgical metastasectomy has a long history of use in oligometastatic disease, with the data demonstrating high local control rates and potential survival advantage.

●●

Completeness of resection is a predictor for improved survival.

●●

Stereotactic ablative body radiation therapy results are promising, with high local control rates and low toxicity reported.

●●

However, the stereotactic ablative body radiation therapy experience is still limited with short duration of follow-up compared with surgery.

●●

Currently no randomized evidence exists to support either surgery or stereotactic ablative body radiation therapy in this setting, and clinical decision-making is based on individual patient and tumor characteristics, and clinical judgment.

Targeted therapy ●●

Molecularly targeted agents that act by blocking signal transduction pathways associated with renal cell carcinoma

tumor growth and angiogenesis have demonstrated higher objective response rates compared with cytokine therapy. ●●

The role of local therapies to the primary tumor or metastases in the era of targeted therapy remains undefined. Randomized trials are ongoing.

Immunotherapy ●●

There is increasing recognition of immunologic effects of high-dose radiation, with ongoing research in this area to try to improve the therapeutic ratio with combined treatment.

Challenges ●●

Local aggressive therapies may not benefit all patients, therefore, it is imperative to identify potentially suitable cases.

●●

Selection of the optimal local therapy should be informed by an understanding of the advantages/disadvantages of the different treatment modalities.

●●

The optimal sequencing or integration of systemic and local therapies in this patient cohort is not yet fully understood.

Future perspective ●●

There is a clear need for well controlled prospective evidence to inform treatment options in patients oligometastatic renal cell carcinoma.

future science group

www.futuremedicine.com

771

Review  Loh, Davis, Martin & Siva improve outcome will arise from combining SABR with other modalities of treatment, ­including ­targeted therapies and immunotherapies. Financial & competing interests disclosure The authors have no relevant affiliations or financial involvement with any organization or entity with a 11 Gupta GP, Massague J. Cancer metastasis:

References Papers of special note have been highlighted as: • of interest; •• of considerable interest 1

Hellman S, Weichselbaum RR. Oligometastases. J. Clin. Oncol. 13(1), 8–10 (1995).

•• The concept of oligometastases was first described here. 2

Macdermed DM, Weichselbaum RR, Salama JK. A rationale for the targeted treatment of oligometastases with radiotherapy. J. Surg. Oncol. 98(3), 202–206 (2008).

3

Weickhardt AJ, Scheier B, Burke JM et al. Local ablative therapy of oligoprogressive disease prolongs disease control by tyrosine kinase inhibitors in oncogene-addicted non-small-cell lung cancer. J. Thorac. Oncol. 7(12), 1807–1814 (2012).

•• Retrospective study suggesting that the approach of using of local ablative therapy for oligoprogressive disease on systemic therapy may prolong disease control. 4

5

6

7

8

9

Christodouleas JP, Marks LB. Analysis of patients with oligometastases undergoing two or more curative-intent stereotactic radiotherapy courses: in regard to Milano et al. (Int J Radiat Oncol Biol Phys 2009;73: 832–837). Int. J. Radiat. Oncol. Biol. Phys. 74(5), 1628; author reply 1628–1629 (2009). Niibe Y, Hayakawa K. Oligometastases and oligo-recurrence: the new era of cancer therapy. Jpn J. Clin. Oncol. 40(2), 107–111 (2010). Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N. Engl. J. Med. 335(12), 865–875 (1996).

building a framework. Cell 127(4), 679–695 (2006). 12 Gerlinger M, Rowan AJ, Horswell S et al.

Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366(10), 883–892 (2012). 13 Kosari F, Parker AS, Kube DM et al. Clear

cell renal cell carcinoma: gene expression analyses identify a potential signature for tumor aggressiveness. Clin. Cancer Res. 11(14), 5128–5139 (2005). 14 Wuttig D, Baier B, Fuessel S et al. Gene

signatures of pulmonary metastases of renal cell carcinoma reflect the disease-free interval and the number of metastases per patient. Int. J. Cancer 125(2), 474–482 (2009). 15 Lussier YA, Xing HR, Salama JK et al.

MicroRNA expression characterizes oligometastasis(es). PLoS ONE 6(12), e28650 (2011). 16 Sohaib SA, Cook G, Allen SD, Hughes M,

Eisen T, Gore M. Comparison of whole-body MRI and bone scintigraphy in the detection of bone metastases in renal cancer. Br. J. Radiol. 82(980), 632–639 (2009). 17 Lawrentschuk N, Davis ID, Bolton DM,

Scott AM. Functional imaging of renal cell carcinoma. Nat. Rev. Urol. 7(5), 258–266 (2010). 18 Oosterwijk-Wakka JC, Boerman OC,

Mulders PF, Oosterwijk E. Application of monoclonal antibody G250 recognizing carbonic anhydrase IX in renal cell carcinoma. Int. J. Mol. Sci. 14(6), 11402–11423 (2013).

Goetz P, Ebinu JO, Roberge D, Zadeh G. Current standards in the management of cerebral metastases. Int. J. Surg. Oncol. 2012, 493426 (2012).

19 Barney JJ. A twelve-year cure following

Eichler AF, Loeffler JS. Multidisciplinary management of brain metastases. Oncologist 12(7), 884–898 (2007).

20 Pfannschmidt J, Hoffmann H, Muley T,

Paget S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8(2), 98–101 (1989).

10 Sluka P, Davis ID. Cell mates: paracrine and

stromal targets for prostate cancer therapy. Nat. Rev. Urol. 10(8), 441–451 (2013).

772

financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

nephrectomy for adenocarcinoma and lobectomy for solitary metastasis. Trans. Am. Assoc. Genitourin. Surg. 37, 189–191 (1945). Krysa S, Trainer C, Dienemann H. Prognostic factors for survival after pulmonary resection of metastatic renal cell carcinoma. Ann. Thorac. Surg. 74(5), 1653–1657 (2002). 21 Hofmann HS, Neef H, Krohe K, Andreev P,

Silber RE. Prognostic factors and survival after pulmonary resection of metastatic renal

Future Oncol. (2014) 10(5)

cell carcinoma. Eur. Urol. 48(1), 77–81; discussion 81–72 (2005). 22 Meimarakis G, Angele M, Staehler M et al.

Evaluation of a new prognostic score (Munich score) to predict long-term survival after resection of pulmonary renal cell carcinoma metastases. Am. J. Surg. 202(2), 158–167 (2011). 23 Murthy SC, Kim K, Rice TW et al. Can we

predict long-term survival after pulmonary metastasectomy for renal cell carcinoma? Ann. Thorac. Surg. 79(3), 996–1003 (2005). 24 Assouad J, Petkova B, Berna P, Dujon A,

Foucault C, Riquet M. Renal cell carcinoma lung metastases surgery: pathologic findings and prognostic factors. Ann. Thorac. Surg. 84(4), 1114–1120 (2007). 25 Baloch KG, Grimer RJ, Carter SR, Tillman

RM. Radical surgery for the solitary bony metastasis from renal-cell carcinoma. J. Bone Joint Surg. Br. 82(1), 62–67 (2000). 26 Fuchs B, Trousdale RT, Rock MG. Solitary

bony metastasis from renal cell carcinoma: significance of surgical treatment. Clin. Orthop. Relat. Res. (431), 187–192 (2005). 27 Jung ST, Ghert MA, Harrelson JM, Scully SP.

Treatment of osseous metastases in patients with renal cell carcinoma. Clin. Orthop. Relat. Res. (409), 223–231 (2003). 28 Lin PP, Mirza AN, Lewis VO et al. Patient

survival after surgery for osseous metastases from renal cell carcinoma. J. Bone Joint Surg. Am. 89(8), 1794–1801 (2007). 29 Szendroi A, Dinya E, Kardos M et al.

Prognostic factors and survival of renal clear cell carcinoma patients with bone metastases. Pathol. Oncol. Res. 16(1), 29–38 (2010). 30 Kozlowski JM. Management of distant

solitary recurrence in the patient with renal cancer. Contralateral kidney and other sites. Urol. Clin. North Am. 21(4), 601–624 (1994). 31 Iesalnieks I, Winter H, Bareck E et al. Thyroid

metastases of renal cell carcinoma: clinical course in 45 patients undergoing surgery. Assessment of factors affecting patients’ survival. Thyroid 18(6), 615–624 (2008). 32 Staehler MD, Kruse J, Haseke N et al. Liver

resection for metastatic disease prolongs survival in renal cell carcinoma: 12-year

future science group

Extracranial oligometastatic renal cell carcinoma: current management & future directions  results from a retrospective comparative analysis. World J. Urol. 28(4), 543–547 (2010).

43 Wersall PJ, Blomgren H, Lax I et al.

Extracranial stereotactic radiotherapy for primary and metastatic renal cell carcinoma. Radiother. Oncol. 77(1), 88–95 (2005).

33 Aloia TA, Adam R, Azoulay D, Bismuth H,

Castaing D. Outcome following hepatic resection of metastatic renal tumors: the Paul Brousse Hospital experience. HPB (Oxford) 8(2), 100–105 (2006). 34 Thelen A, Jonas S, Benckert C et al. Liver

resection for metastases from renal cell carcinoma. World J. Surg. 31(4), 802–807 (2007). 35 Zerbi A, Ortolano E, Balzano G, Borri A,

Beneduce AA, Di Carlo V. Pancreatic metastasis from renal cell carcinoma: which patients benefit from surgical resection? Ann. Surg. Oncol. 15(4), 1161–1168 (2008). 36 Reddy S, Wolfgang CL. The role of surgery in

the management of isolated metastases to the pancreas. Lancet Oncol. 10(3), 287–293 (2009). 37 Alt AL, Boorjian SA, Lohse CM, Costello

BA, Leibovich BC, Blute ML. Survival after complete surgical resection of multiple metastases from renal cell carcinoma. Cancer 117(13), 2873–2882 (2011). •

Retrospective study demonstrates a survival advantage in patients who had a complete resection of their metastatic renal cell carcinoma disease over those who had an incomplete resection, even in the presence of other less favorable prognostic factors.

38 Siva S, Macmanus M, Ball D. Stereotactic

radiotherapy for pulmonary oligometastases: a systematic review. J. Thorac. Oncol. 5(7), 1091–1099 (2010). 39 Ning S, Trisler K, Wessels BW, Knox SJ.

Radiobiologic studies of radioimmunotherapy and external beam radiotherapy in vitro and in vivo in human renal cell carcinoma xenografts. Cancer 80(12 Suppl.), 2519–2528 (1997). 40 Ranck MC, Golden DW, Corbin KS et al.

Stereotactic body radiotherapy for the treatment of oligometastatic renal cell carcinoma. Am. J. Clin. Oncol. 36(6), 589–595 (2013). 41 Zelefsky MJ, Greco C, Motzer R et al. Tumor

control outcomes after hypofractionated and single-dose stereotactic image-guided intensity-modulated radiotherapy for extracranial metastases from renal cell carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 82(5), 1744–1748 (2012). 42 Svedman C, Sandstrom P, Pisa P et al.

A prospective Phase II trial of using extracranial stereotactic radiotherapy in primary and metastatic renal cell carcinoma. Acta Oncol. 45(7), 870–875 (2006).

future science group



Demonstrates high local control rates for stereotactic ablative body radiation therapy in primary and metastatic renal cell carcinoma with low toxicity rates. Survival was longer in patients with less than three metastases compared with those with more than three.

44 Teh B, Bloch C, Galli-Guevara M et al. The

treatment of primary and metastatic renal cell carcinoma (RCC) with image-guided stereotactic body radiation therapy (SBRT). Biomed. Imaging Interv. J. 3(1), e6 (2007). 45 Gilson B LG, Qian G, Fastaia M, Cangiane

L. Hypo-fractionated stereotactic extracranial radiosurgery (hfsr) for primary and metastatic renal cell carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 66, S349–S349 (2006). 46 Balagamwala EH, Angelov L, Koyfman SA

et al. Single-fraction stereotactic body radiotherapy for spinal metastases from renal cell carcinoma. J. Neurosurg. Spine 17(6), 556–564 (2012). 47 Nguyen QN, Shiu AS, Rhines LD et al.

Management of spinal metastases from renal cell carcinoma using stereotactic body radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 76(4), 1185–1192 (2010). 48 Gerszten PC, Burton SA, Ozhasoglu C et al.

Stereotactic radiosurgery for spinal metastases from renal cell carcinoma. J. Neurosurg. Spine 3(4), 288–295 (2005). 49 Salama JK, Hasselle MD, Chmura SJ et al.

Stereotactic body radiotherapy for multisite extracranial oligometastases: final report of a dose escalation trial in patients with 1 to 5 sites of metastatic disease. Cancer 118(11), 2962–2970 (2012). 50 Beitler JJ, Makara D, Silverman P, Lederman

G. Definitive, high-dose-per-fraction, conformal, stereotactic external radiation for renal cell carcinoma. Am. J. Clin. Oncol. 27(6), 646–648 (2004). 51 Siva S, Pham D, Gill S, Corcoran NM,

Foroudi F. A systematic review of stereotactic radiotherapy ablation for primary renal cell carcinoma. BJU Int. 110(11 Pt B), e737–e743 (2012). 52 Motzer RJ, Hutson TE, Tomczak P et al.

Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 356(2), 115–124 (2007). 53 Sternberg CN, Davis ID, Mardiak J et al.

Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized

Review

Phase III trial. J. Clin. Oncol. 28(6), 1061–1068 (2010). 54 Motzer RJ, Hutson TE, Cella D et al.

Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N. Engl. J. Med. 369(8), 722–731 (2013). 55 Hudes G, Carducci M, Tomczak P et al.

Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 356(22), 2271–2281 (2007). 56 Escudier B, Pluzanska A, Koralewski P et al.

Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind Phase III trial. Lancet 370(9605), 2103–2111 (2007). 57 Rini BI, Halabi S, Rosenberg JE et al.

Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. J. Clin. Oncol. 28(13), 2137–2143 (2010). 58 Escudier B, Szczylik C, Hutson TE et al.

Randomized Phase II trial of first-line treatment with sorafenib versus interferon alfa-2a in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 27(8), 1280–1289 (2009). 59 Motzer RJ, Escudier B, Oudard S et al.

Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled Phase III trial. Lancet 372(9637), 449–456 (2008). 60 Rini BI, Escudier B, Tomczak P et al.

Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised Phase 3 trial. Lancet 378(9807), 1931–1939 (2011). 61 Escudier B, Eisen T, Stadler WM et al.

Sorafenib in advanced clear-cell renal-cell carcinoma. N. Engl. J. Med. 356(2), 125–134 (2007). 62 Motzer RJ, Nosov D, Eisen T et al.

Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a Phase III trial. J. Clin. Oncol. 31(30), 3791–3799 (2013). 63 Mickisch GH, Garin A, Van Poppel H et al.

Radical nephrectomy plus interferon-alfabased immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet 358(9286), 966–970 (2001). 64 Flanigan RC, Salmon SE, Blumenstein BA

et al. Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell cancer. N. Engl. J. Med. 345(23), 1655–1659 (2001).

www.futuremedicine.com

773

Review  Loh, Davis, Martin & Siva 65 Choueiri TK, Xie W, Kollmannsberger C

et al. The impact of cytoreductive nephrectomy on survival of patients with metastatic renal cell carcinoma receiving vascular endothelial growth factor targeted therapy. J. Urol. 185(1), 60–66 (2011).

76 Postow MA, Callahan MK, Barker CA et al.

Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 366(10), 925–931 (2012). 77 Siva S, Macmanus MP, Martin RF, Martin

OA. Abscopal effects of radiation therapy: a clinical review for the radiobiologist. Cancer Lett. doi:10.1016/j.canlet.2013.09.018 (2013) (Epub ahead of print).).

66 Warren M, Venner PM, North S et al. A

population-based study examining the effect of tyrosine kinase inhibitors on survival in metastatic renal cell carcinoma in Alberta and the role of nephrectomy prior to treatment. Can. Urol. Assoc. J. 3(4), 281–289 (2009).

78 Siva S, Callahan J, Macmanus MP, Martin O,

Hicks RJ, Ball DL. Abscopal [corrected] effects after conventional and stereotactic lung irradiation of non-small-cell lung cancer. J. Thorac. Oncol. 8(8), e71–e72 (2013).

67 Barbastefano J, Garcia JA, Elson P et al.

Association of percentage of tumour burden removed with debulking nephrectomy and progression-free survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. BJU Int. 106(9), 1266–1269 (2010).

79 Wersall PJ, Blomgren H, Pisa P, Lax I,

Kalkner KM, Svedman C. Regression of non-irradiated metastases after extracranial stereotactic radiotherapy in metastatic renal cell carcinoma. Acta Oncol. 45(4), 493–497 (2006).

68 ClinicalTrials.gov. Randomized Phase III

Trial Evaluating The Importance Of Nephrectomy In Patients Presenting With Metastatic Renal Cell Carcinoma Treated With Sunitinib. http://clinicaltrials.gov/show/NCT00930033 69 ClinicalTrials.gov. Randomized Phase III

Trial Comparing Immediate Versus Deferred Nephrectomy in Patients With Synchronous Metastatic Renal Cell Carcinoma. http://clinicaltrials.gov/show/NCT01099423 70 Johannsen M, Staehler M, Ohlmann CH et al.

Outcome of treatment discontinuation in patients with metastatic renal cell carcinoma and no evidence of disease following targeted therapy with or without metastasectomy. Ann. Oncol. 22(3), 657–663 (2011). 71 Chapin BF, Delacroix SE Jr, Culp SH et al.

80 Formenti SC, Demaria S. Combining

radiotherapy and cancer immunotherapy: a paradigm shift. J. Natl Cancer Inst. 105(4), 256–265 (2013). 81 Matzinger P. Tolerance, danger, and the

extended family. Annu. Rev. Immunol. 12(1), 991–1045 (1994). 82 Matzinger P. The danger model: a renewed

sense of self. Science 296(5566), 301 (2002). 83 Lugade AA, Sorensen EW, Gerber SA, Moran

JP, Frelinger JG, Lord EM. Radiation-induced IFN-γ production within the tumor microenvironment influences antitumor immunity. J. Immunol. 180(5), 3132–3139 (2008). 84 Lee Y, Auh SL, Wang Y et al. Therapeutic

effects of ablative radiation on local tumor require CD8 + T cells: changing strategies for cancer treatment. Blood 114(3), 589–595 (2009).

Safety of presurgical targeted therapy in the setting of metastatic renal cell carcinoma. Eur. Urol. 60(5), 964–971 (2011). 72 Powles T, Kayani I, Blank C et al. The safety

and efficacy of sunitinib before planned nephrectomy in metastatic clear cell renal cancer. Ann. Oncol. 22(5), 1041–1047 (2011). 73 Karam JA, Rini BI, Varella L et al.

Metastasectomy after targeted therapy in patients with advanced renal cell carcinoma. J. Urol. 185(2), 439–444 (2011). 74 Tong CC, Ko EC, Sung MW et al. Phase II

trial of concurrent sunitinib and imageguided radiotherapy for oligometastases. PLoS ONE 7(6), e36979 (2012). 75 Bockorny B, Dasanu CA. Intrinsic immune

alterations in renal cell carcinoma and emerging immunotherapeutic approaches. Expert Opin Biol. Ther. 13(6), 911–925 (2013).

774



Through experiments using animal models, the authors demonstrated that reduction of tumor burden after ablative radiation therapy depended largely on T-cell responses, and the ablative radiation therapy immune responses are greatly amplified by local immunotherapy.

85 Drake CG, Lipson EJ, Brahmer JR. Breathing

new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat. Rev. Clin. Oncol. 11(1), 24–37 (2013). 86 Pardoll DM. The blockade of immune

checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12(4), 252–264 (2012). 87 Topalian SL, Hodi FS, Brahmer JR et al.

Safety, activity, and immune correlates of

Future Oncol. (2014) 10(5)

anti-PD-1 antibody in cancer. N. Engl. J. Med. 366(26), 2443–2454 (2012). •• Demonstrates that anti-PD-1 antibody produced objective responses with durability in one in four to one in five patients of various primary tumors including renal cell carcinoma, and with an adverse event profile that suggests feasibility of outpatient administration with minimal supportive care. 88 Wolchok JD, Kluger H, Callahan MK et al.

Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 369(2), 122–133 (2013). 89 Weber JS, Kudchadkar RR, Yu B et al. Safety,

efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J. Clin. Oncol. 31(34), 4311–4318 (2013). 90 Hamid O, Robert C, Daud A et al. Safety and

tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 369(2), 134–144 (2013). 91 ClinicalTrials.gov. A Randomized, Open-

Label, Phase 3 Study of Nivolumab (BMS-936558) vs. Everolimus in Subjects With Advanced or Metastatic Clear-Cell Renal Cell Carcinoma Who Have Received Prior Anti-Angiogenic Therapy. http://clinicaltrials.gov/show/NCT01668784 92 Yang JC, Hughes M, Kammula U et al.

Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J. Immunother. 30(8), 825–830 (2007). 93 Ozao-Choy J, Ma G, Kao J et al. The novel

role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res. 69(6), 2514–2522 (2009). 94 ClinicalTrials.gov. A Phase 1 Study of

Nivolumab (BMS-936558) Plus Sunitinib, Pazopanib or Ipilimumab in Subjects With Metastatic Renal Cell Carcinoma. http://clinicaltrials.gov/show/NCT01472081 95 Seung SK, Curti BD, Crittenden M et al.

Phase 1 study of stereotactic body radiotherapy and interleukin-2–tumor and immunological responses. Sci. Transl. Med. 4(137), 137ra174 (2012). 96 Masucci GV, Wersall P, Kiessling R,

Lundqvist A, Lewensohn R. Stereotactic ablative radio therapy (SABR) followed by immunotherapy a challenge for individualized treatment of metastatic solid tumours. J. Transl. Med. 10, 104 (2012).

future science group