Search

Recent Articles

IJMPO—A Journey of a Thousand Miles

Author : Padmaj S. Kulkarni

Coronavirus Disease 2019 Treatment—T-Cells Hold the Key in Severe Cases

Author : Kunal Das, Nitika Agrawal, Mansi Kala, Rakhee Khanduri

Why Is China Importing COVID-19 Vaccine Now?

Author : Purvish M. Parikh

Oligometastatic breast cancer: A mini review

CC BY-NC-ND 4.0 · Indian J Med Paediatr Oncol 2014; 35(03): 203-206

DOI: DOI: 10.4103/0971-5851.142035

Abstract

With few exceptions such as germ cell tumors, trophoblastic neoplasms and colonic cancers, metastatic solid tumors are considered largely incurable. It is increasingly appreciated that oligometaststic cancer differs from multi-metastatic disease in prognosis and survival. Oligometastatic breast cancer (OMBC) is, therefore, sometimes considered as an intermediate biological state between localized and widely metastatic disease. There is no strict definition of OMBC with studies using different criteria. Treatment of OMBC is still controversial in view of sparse data that is retrospective. However, there is an increasing shift toward individualized, multidisciplinary management of OMBC with the intent to cure some patients. This article will concisely review the subject of OMBC from points of view of biology and practical management recommendations.



Publication History

Article published online:
19 July 2021

© 2014. Indian Society of Medical and Paediatric Oncology. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/.)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

We recommend

  1. CyberKnife Stereotactic Ablative Radiotherapy for Recurrent or Oligometastatic Gynecological Cancers
    Tejinder Kataria et al., South Asian Journal of Cancer, 2021
  2. Newly Diagnosed Sellar Tumors in Patients with Cancer. Advantages of Surgical Intervention Over Empiric Radiotherapy
    Carlos R. Goulart et al., Journal of Pediatric Biochemistry
  3. Radioembolization of Secondary Hepatic Malignancies
    Barbara Manchec et al., Seminars in Interventional Radiology, 2021
  4. Surgical Management of Intracerebral Metastases
    Christopher G. Kalhorn et al., Seminars in Neurosurgery, 2000
  5. Circulating and Disseminated Tumor Cells in Breast Carcinoma
    Malgorzata Banys-Paluchowski et al., Geburtshilfe und Frauenheilkunde, 2019

Abstract

With few exceptions such as germ cell tumors, trophoblastic neoplasms and colonic cancers, metastatic solid tumors are considered largely incurable. It is increasingly appreciated that oligometaststic cancer differs from multi-metastatic disease in prognosis and survival. Oligometastatic breast cancer (OMBC) is, therefore, sometimes considered as an intermediate biological state between localized and widely metastatic disease. There is no strict definition of OMBC with studies using different criteria. Treatment of OMBC is still controversial in view of sparse data that is retrospective. However, there is an increasing shift toward individualized, multidisciplinary management of OMBC with the intent to cure some patients. This article will concisely review the subject of OMBC from points of view of biology and practical management recommendations.

Keywords: Oligometastastasisbreast cancercure

INTRODUCTION

Metastatic breast cancer (MBC) is generally considered incurable but advances in treatment coupled with stage migration due to improved imaging and diagnostics has resulted in significantly longer survival in the last few years.[,] OMBC is a subset of MBC with limited number and sites of metastasis and constitutes as high as 20% of all MBCs.[] There is increasing evidence that prolonged disease control is possible in patients with OMBC when treated with aggressive multidisciplinary management.[,] It is still debatable whether long term survival in this subset is due to selection of patients whose tumors have indolent disease biology or to effects of therapy. The sparse data, heterogeneity of disease biology and absence of randomized trials make treatment recommendations less evidence based.

BIOLOGICAL BASIS OF OLIGOMETASTATIC BREAST CANCER

Metastasis is a series of steps involving complex interactions between the tumor cells, microenvironment and host. Genetic, epigenetic and host immune processes contribute to the balance that is permissive of metastasis. Genes that are responsible for efficient metastasis have been recently characterized as initiator, progression and virulent genes.[] Metastasis “initiation” genes provide selective survival or growth advantage to the primary tumor cells; “progression” genes are those that facilitate rate-limiting functions in colonization; and ‘virulence’ genes are those that provide an advantage in metastatic colonization and growth but not necessarily in the primary tumor. Deficiency in one or more of these pathways could potentially lead to impaired but not completely abrogated metastatic capacity in tumor cells and could constitute the biological substrate of oligometastatic disease. In this model, the rationale for eradicative treatment of oligometastatic disease would be premised on preventing further clonal evolution leading to the acquisition of full potential for widespread metastases. Evidence of evolution of oligometastasis phenotype comes from various clinical and preclinical studies. Fidler et al. showed differential metastatic ability of different tumor-cell clones derived from B16F1 melanoma lines to colonize the lungs in syngenic mice.[] Similarly, differential metastatic activity and clonal heterogeneity have been demonstrated in other cell lines.[,]

Oligometastatic phenotype and behavior are generally consistent with cell lines of low malignant potential and their in vivo counterparts could potentially give rise to oligometastatic disease. Genomic instability was initially recognized as a hallmark of cancer development but in last few years it has also been shown to be involved in cancer progression and metastasis. Yachida et al. demonstrated the accumulation of sequential hierarchical genetic changes from primary pancreatic tumor formation through its metastatic progression.[] This temporal acquisition of genetic changes suggests that some oligometastatic tumors that have less than fully evolved metastatic repertoire could be amenable to potentially curative treatment consistent with their biology. Recent improvements in sensitivity and sophistication of imaging technology have led to increasing detection of oligometastatic disease, some of it in evolution to full metastatic picture and some destined for indolent, non-progressive behavior.[] The continuing challenge is to discover biomarkers that will segregate, at diagnosis, the preceding groups of patients with oligometastatic disease, with obvious therapeutic implications.

TREATMENT OF OLIGOMETASTATIC BREAST CANCER

These patients can be clinically divided into those with upfront presentation as OMBC and those with OMBC at relapse. The goals of treatment in MBC are conventionally considered to be increased survival and better quality of life. However, in subsets of patients, such as those with OMBC, the surrogate goals are increasingly changing to complete remission (CR) at clinical or cellular levels. In this context, because of lack of proven progression to clinical disease in all cases, cellular CR cannot be considered to be a clinically relevant end point.[]

Chemotherapy as a single modality

There is no study that has systematically evaluated the chemotherapy alone for patients with OMBC. Greenberg et al. retrospectively reviewed 1581 MBC patients, mostly chemotherapy naïve, who were treated with anthracycline and alkylating agents based regimens of whom 1293 (82%) had 1-3 metastatic sites.[] After completion of treatment, 3.1% of all patients remained in CR for more than 5-year of whom 92% had oliogometastatic (1-2 sites) disease at baseline. This suggests a somewhat higher propensity for OMBC to attain long term remissions and functional cures.[] This study along with others suggests that patients with low tumor burden metastatic disease are likelier to achieve CR, in turn implying that long term survival was related to low tumor burden/OMBC.[] It remains unclear whether long term survival is due to chemotherapy or favorable disease biology or their interaction. A randomized trial addressing this issue will be difficult to implement because of obvious reasons.

High-dose chemotherapy for oligometastatic breast cancer

High-dose chemotherapy (HDCT) followed by autologous peripheral blood progenitor cell rescue has been compared with conventional chemotherapy in patients with MBC in many trials, but the approach remains controversial. The Cochrane review included six randomized controlled trials (RCTs) demonstrating a significantly improved event-free survival (EFS) at 3, 4, and 5 years after treatment with HDCT, but no overall survival (OS) benefit.[] Similarly, a recent meta-analysis of 15 trials that included data on 6210 patients showed significant EFS benefit but none in OS, in favor of HDCT.[]

Data for OMBC treated with HDCT is sparse. Nieto et al. reported a prospective study of 60 OMBC patients who received induction chemotherapy followed by local treatment followed by hematopoietic stem cell transplantation (HSCT).[] The 5-year relapse free survival was 52% in this population. Bojko et al., in a similar small prospective study involving OMBC patients treated with induction chemotherapy followed by local treatment followed by HSCT, reported a 5-year progression free survival (PFS) of 27%.[] The difference in outcomes between the two preceding studies could be explained by inclusion of patients with liver metastases (29%) in the second study. Although the results are encouraging, based on these small nonrandomized studies no recommendations can be made regarding the use of HSCT in OMBC patients. Future studies should aim at evaluating the molecular characteristics of patients who may benefit from this treatment.

Surgery of the primary site in oligometastatic breast cancer

The potential advantages of removing the primary tumor in OMBC is elimination of a potential source of further metastatic seeding, restoration of immune competence and reduction in chemoresistance by reducing the number of clones.[] Arguments against removal of the primary tumor include Fisher's mouse experimental model which suggested increase in the proliferation of distant tumor foci (“metastasis”) associated with the removal of primary tumor masses in different tumor types.[]

Until recently there were several retrospective series that showed benefit of local treatment of the primary tumor but these studies included selected patients with better performance status, less advanced primary tumor and lower disease burden. It is difficult to ascribe the outcomes to local treatment versus disease biology in these reports.[]

Badwe et al. conducted a RCT to address this issue.[] In the overall study population (N = 350) of patients with MBC who were responding to anthracycline based chemotherapy, they found no benefit of loco-regional therapy (LRT) in terms of OS. In a preplanned subset of patients with oligometastatic (≤3) disease there was no difference in OS between the LRT and no LRT arms. Until the presentation of further randomized results, this remains the best evidence on this question and routine surgery for the primary tumor cannot be recommended in OMBC.

Role of surgery/local therapy of metastatic sites

Again, there is sparse evidence for metastatectomy in patients with OMBC. The largest retrospective data comes from International Registry of Lung Metastasis, which was established in 1991 to collect the experience of curative intent surgery for pulmonary metastases. Among breast cancer patients (N = 467) who underwent metastatectomy, the median survival was 35 months with 15-year survival of 18%.[] Being retrospective and nonrandomized this data suffers from the same biases that were earlier pointed out for local therapy in such cases.[] Although evidence remains inconclusive about its therapeutic value, pulmonary metastatectomy does have a useful role in the confirmation of diagnosis in some patients. Rena et al. evaluated a series of 79 consecutive patients who underwent surgery for solitary pulmonary nodule after a curative resection for breast cancer.[] Histopathological evaluation of the resected specimen revealed primary lung cancer in 38 patients, pulmonary metastasis from breast cancer in 27 and benign conditions in 14.

Unlike pulmonary resection, liver resection is practiced less often but multiple retrospective analyses in highly selected group of patients show favorable long term survival. Other techniques of local control like sterotactic body radiotherapy and radiofrequency ablation are being increasingly used, but their role is not yet clear.[,]

Thus, metastatectomy in OMBC for therapeutic benefit is not considered standard of care but may be undertaken in individual cases especially to rule out other diagnoses.[]

Role of adjuvant or pseudoadjuvant systemic therapy

Borner et al. reported the first randomized trial in OMBC wherein patients (N = 167) with ‘good-risk’ isolated locoregional recurrence (ILRR) were randomized, after local treatment, to receive tamoxifen or not.[] Tamoxifen improved the median disease free survival (DFS) from 26 to 82 months (P = 0.007) but OS was not significantly increased, perhaps because of small sample size and short follow-up.

Chemotherapy in the so called “pseudoadjuvant” setting has been supported mainly by phase II trials. The largest data is from M. D. Anderson Cancer Center which published the outcome of patients in four phase II trials utilizing combined modality for the treatment of isolated recurrences.[] Patients received local therapy with curative intent and efficacy of “adjuvant” chemotherapy in subjects with clinical CR was evaluated. Three of the 4 studies used doxorubicin based chemotherapy and after a median follow-up of 121.5 months the estimated 20-year DFS and OS were both 26% in these studies. With a shorter median followup in the docetaxel based trial, the DFS was 58%. However, potential selection bias and inclusion of anthracycline and taxane naïve patients in these studies makes their results less generalizable.

The recently published CALOR trial also sheds light on the benefit of chemotherapy in completely resected ILRR of breast cancer.[] Patients (N = 162) were randomized to chemotherapy (N = 85) versus no chemotherapy arm (N = 77) and a choice of chemotherapy was left to investigator discretion. Chemotherapy reduced both distant and second local failures, and 5-year DFS was 69% in the chemotherapy group when compared to 57% in no chemotherapy group (P = 0.046). In a prespecified subgroup analysis, adjuvant chemotherapy seemed to be significantly more effective in women with estrogen-receptor-negative ILRR patients assigned to chemotherapy for estrogen-receptor-negative ILRR, but the interaction test was not significant. The authors concluded that ‘adjuvant’ chemotherapy could be recommended in completely resected ILRR, especially if the tumor was ER negative.

Role of neoadjuvant like chemotherapy 

Extrapolating from non-MBC the option of upfront chemotherapy followed by local therapy in responding OMBC patients could be explored. The potential advantage of this strategy could be exclusion of patients with chemoresistant disease (and potentially poor outcomes) from local therapy. Kobayashi et al. have recently published a retrospective analysis of patients (N = 75) treated with this strategy and the experimental arm of Badwe et al. also utilized the same treatment plan.[,] In the former study, at a median follow-up of 103 months, the estimated 10- and 20-year OS rates were 59.2% and 34.1% respectively. These results can, at least partly, be explained by selection of good prognostic patients (60% with metastasis in one organ and 40% chemotherapy naïve).

CONCLUSION

Oligometastatic breast cancer is a subgroup of patients with MBC who have good long term survival raising the tantalizing possibility of “functional cure.” However, it is still uncertain whether these ‘cures’ are due to selection of patients with favorable disease biology versus ‘aggressive’ local or systemic therapeutic interventions. The increasing sensitivity and sophistication in detection of metastatic disease is likely to increase the number of OMBC patients. Depending on a number of host and tumor characteristics, at least some of these patients are candidates for multi-modality therapy. Archived tumor tissue from long term OMBC survivors is likely to be a valuable resource in dissecting the biology of these tumors compared to patients with multi-metastatic disease. Collaborative efforts at clinical, pathological and molecular data collection are therefore very much in order.

Footnotes

Source of Support: Nil.

Conflict of Interest: None declared.

References

  1. Mauri D, Polyzos NP, Salanti G, Pavlidis N, Ioannidis JP. Multiple-treatments meta-analysis of chemotherapy and targeted therapies in advanced breast cancer. J Natl Cancer Inst 2008;100:1780-91.
  2. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol 1995;13:8-103.
  3. Jain SK, Dorn PL, Chmura SJ, Weichselbaum RR. Incidence and implications of oligometastatic breast cancer. J Clin Oncol 2012;30. [Suppl; abstr e11512].
  4. Hanrahan EO, Broglio KR, Buzdar AU, Theriault RL, Valero V, Cristofanilli M, et al. Combined-modality treatment forisolated recurrences of breast carcinoma: Update on 30 years of experience at the University of Texas M.D. Anderson Cancer Center and assessment of prognostic factors. Cancer 2005;104:1158-71.
  5. Kobayashi T, Ichiba T, Sakuyama T, Arakawa Y, Nagasaki E, Aiba K, et al. Possible clinical cure of metastatic breast cancer: Lessons from our 30-year experience with oligometastatic breast cancer patients and literature review. Breast Cancer 2012;19:218-37.
  6. Gupta GP, Massagué J. Cancer metastasis: Building a framework. Cell 2006;127:679-95.
  7. Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within a malignant tumor. Science 1977;197:893-5.
  8. Li Y, Tang ZY, Ye SL, Liu YK, Chen J, Xue Q, et al. Establishment of cell clones with different metastatic potential from the metastatic hepatocellular carcinoma cell line MHCC97. World J Gastroenterol 2001;7:630-6.
  9. Shindo-Okada N, Takeuchi K, Nagamachi Y. Establishment of cell lines with high-and low-metastatic potential from PC-14 human lung adenocarcinoma. Jpn J Cancer Res 2001;92:174-83.
  10. Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114-7.
  11. Corbin KS, Hellman S, Weichselbaum RR. Extracranial oligometastases: A subset of metastases curable with stereotactic radiotherapy. J Clin Oncol 2013;31:1384-90.
  12. Mansi JL, Gogas H, Bliss JM, Gazet JC, Berger U, Coombes RC. Outcome of primary-breast-cancer patients with micrometastases: A long-term follow-up study. Lancet 1999;354:197-202.
  13. Greenberg PA, Hortobagyi GN, Smith TL, Ziegler LD, Frye DK, Buzdar AU. Long-term follow-up of patients with complete remission following combination chemotherapy for metastatic breast cancer. J Clin Oncol 1996;14:2197-205.
  14. Swenerton KD, Legha SS, Smith T, Hortobagyi GN, Gehan EA, Yap HY, et al. Prognostic factors in metastatic breast cancer treated with combination chemotherapy. Cancer Res 1979;39:1552-62.
  15. Farquhar C, Basser R, Marjoribanks J, Lethaby A. High dose chemotherapy and autologous bone marrow or stem cell transplantation versus conventional chemotherapy for women with early poor prognosis breast cancer. The Cochrane Database of Systematic Reviews 2003; CD003139. DOI: 10.1002/14651858.
  16. Berry DA, Ueno NT, Johnson MM, Lei X, Caputo J, Rodenhuis S, et al. High-dose chemotherapy with autologous stem-cell support as adjuvant therapy in breast cancer: Overview of 15 randomized trials. J Clin Oncol 2011;29:3214-23.
  17. Nieto Y, Nawaz S, Jones RB, Shpall EJ, Cagnoni PJ, McSweeney PA, et al. Prognostic model for relapse after high-dose chemotherapy with autologous stem-cell transplantation for stage IV oligometastatic breast cancer. J Clin Oncol 2002;20:707-18.
  18. Bojko P, Welt A, Schleucher R, Borquez D, Scheulen ME, Vanhoefer U, et al. High-dose chemotherapy with autologous stem cell transplantation in patients with oligometastatic breast cancer. Bone Marrow Transplant 2004;34:637-43.
  19. Pagani O, Senkus E, Wood W, Colleoni M, Cufer T, Kyriakides S, et al. International guidelines for management of metastatic breast cancer: Can metastatic breast cancer be cured? J Natl Cancer Inst 2010;102:456-63.
  20. Fisher B, Gunduz N, Coyle J, Rudock C, Saffer E. Presence of a growth-stimulating factor in serum following primary tumor removal in mice. Cancer Res 1989;49:1996-2001.
  21. Olson JA Jr, Marcom PK. Benefit or bias? The role of surgery to remove the primary tumor in patients with metastatic breast cancer. Ann Surg 2008;247:739-40.
  22. Badwe R, Parmar V, Hawaldar R, Nair N, Kaushik R, Siddique S, et al. Surgical removal of primary breast tumor and axillary lymph nodes in women with metastatic breast cancer at first presentation: A randomized controlled trial. [Abstract S2-02]. Presented at: 2013 San Antonio Breast Cancer Symposium; December 10-14, 2013; San Antonio, TX.
  23. Friedel G, Pastorino U, Ginsberg RJ, Goldstraw P, Johnston M, Pass H, et al. Results of lung metastasectomy from breast cancer: Prognostic criteria on the basis of 467 cases of the International Registry of Lung Metastases. Eur J Cardiothorac Surg 2002;22:335-44.
  24. Rena O, Papalia E, Ruffini E, Filosso PL, Oliaro A, Maggi G, et al. The role of surgery in the management of solitary pulmonary nodule in breast cancer patients. Eur J Surg Oncol 2007;33:546-50.
  25. Welter S, Jacobs J, Krbek T, Tötsch M, Stamatis G. Pulmonary metastases of breast cancer. When is resection indicated? Eur J Cardiothorac Surg 2008;34:1228-34.
  26. Borner M, Bacchi M, Goldhirsch A, Greiner R, Harder F, Castiglione M, et al. First isolated locoregional recurrence following mastectomy for breast cancer: Results of a phase III multicenter study comparing systemic treatment with observation after excision and radiation. Swiss Group for Clinical Cancer Research. J Clin Oncol 1994;12:2071-7.
  27. Aebi S, Gelber S, Anderson SJ, Láng I, Robidoux A, Martín M, et al. Chemotherapy for isolated locoregional recurrence of breast cancer (CALOR): A randomised trial. Lancet Oncol 2014;15:156-63.

References

  1. Mauri D, Polyzos NP, Salanti G, Pavlidis N, Ioannidis JP. Multiple-treatments meta-analysis of chemotherapy and targeted therapies in advanced breast cancer. J Natl Cancer Inst 2008;100:1780-91.
  2. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol 1995;13:8-103.
  3. Jain SK, Dorn PL, Chmura SJ, Weichselbaum RR. Incidence and implications of oligometastatic breast cancer. J Clin Oncol 2012;30. [Suppl; abstr e11512].
  4. Hanrahan EO, Broglio KR, Buzdar AU, Theriault RL, Valero V, Cristofanilli M, et al. Combined-modality treatment forisolated recurrences of breast carcinoma: Update on 30 years of experience at the University of Texas M.D. Anderson Cancer Center and assessment of prognostic factors. Cancer 2005;104:1158-71.
  5. Kobayashi T, Ichiba T, Sakuyama T, Arakawa Y, Nagasaki E, Aiba K, et al. Possible clinical cure of metastatic breast cancer: Lessons from our 30-year experience with oligometastatic breast cancer patients and literature review. Breast Cancer 2012;19:218-37.
  6. Gupta GP, Massagué J. Cancer metastasis: Building a framework. Cell 2006;127:679-95.
  7. Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within a malignant tumor. Science 1977;197:893-5.
  8. Li Y, Tang ZY, Ye SL, Liu YK, Chen J, Xue Q, et al. Establishment of cell clones with different metastatic potential from the metastatic hepatocellular carcinoma cell line MHCC97. World J Gastroenterol 2001;7:630-6.
  9. Shindo-Okada N, Takeuchi K, Nagamachi Y. Establishment of cell lines with high-and low-metastatic potential from PC-14 human lung adenocarcinoma. Jpn J Cancer Res 2001;92:174-83.
  10. Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114-7.
  11. Corbin KS, Hellman S, Weichselbaum RR. Extracranial oligometastases: A subset of metastases curable with stereotactic radiotherapy. J Clin Oncol 2013;31:1384-90.
  12. Mansi JL, Gogas H, Bliss JM, Gazet JC, Berger U, Coombes RC. Outcome of primary-breast-cancer patients with micrometastases: A long-term follow-up study. Lancet 1999;354:197-202.
  13. Greenberg PA, Hortobagyi GN, Smith TL, Ziegler LD, Frye DK, Buzdar AU. Long-term follow-up of patients with complete remission following combination chemotherapy for metastatic breast cancer. J Clin Oncol 1996;14:2197-205.
  14. Swenerton KD, Legha SS, Smith T, Hortobagyi GN, Gehan EA, Yap HY, et al. Prognostic factors in metastatic breast cancer treated with combination chemotherapy. Cancer Res 1979;39:1552-62.
  15. Farquhar C, Basser R, Marjoribanks J, Lethaby A. High dose chemotherapy and autologous bone marrow or stem cell transplantation versus conventional chemotherapy for women with early poor prognosis breast cancer. The Cochrane Database of Systematic Reviews 2003; CD003139. DOI: 10.1002/14651858.
  16. Berry DA, Ueno NT, Johnson MM, Lei X, Caputo J, Rodenhuis S, et al. High-dose chemotherapy with autologous stem-cell support as adjuvant therapy in breast cancer: Overview of 15 randomized trials. J Clin Oncol 2011;29:3214-23.
  17. Nieto Y, Nawaz S, Jones RB, Shpall EJ, Cagnoni PJ, McSweeney PA, et al. Prognostic model for relapse after high-dose chemotherapy with autologous stem-cell transplantation for stage IV oligometastatic breast cancer. J Clin Oncol 2002;20:707-18.
  18. Bojko P, Welt A, Schleucher R, Borquez D, Scheulen ME, Vanhoefer U, et al. High-dose chemotherapy with autologous stem cell transplantation in patients with oligometastatic breast cancer. Bone Marrow Transplant 2004;34:637-43.
  19. Pagani O, Senkus E, Wood W, Colleoni M, Cufer T, Kyriakides S, et al. International guidelines for management of metastatic breast cancer: Can metastatic breast cancer be cured? J Natl Cancer Inst 2010;102:456-63.
  20. Fisher B, Gunduz N, Coyle J, Rudock C, Saffer E. Presence of a growth-stimulating factor in serum following primary tumor removal in mice. Cancer Res 1989;49:1996-2001.
  21. Olson JA Jr, Marcom PK. Benefit or bias? The role of surgery to remove the primary tumor in patients with metastatic breast cancer. Ann Surg 2008;247:739-40.
  22. Badwe R, Parmar V, Hawaldar R, Nair N, Kaushik R, Siddique S, et al. Surgical removal of primary breast tumor and axillary lymph nodes in women with metastatic breast cancer at first presentation: A randomized controlled trial. [Abstract S2-02]. Presented at: 2013 San Antonio Breast Cancer Symposium; December 10-14, 2013; San Antonio, TX.
  23. Friedel G, Pastorino U, Ginsberg RJ, Goldstraw P, Johnston M, Pass H, et al. Results of lung metastasectomy from breast cancer: Prognostic criteria on the basis of 467 cases of the International Registry of Lung Metastases. Eur J Cardiothorac Surg 2002;22:335-44.
  24. Rena O, Papalia E, Ruffini E, Filosso PL, Oliaro A, Maggi G, et al. The role of surgery in the management of solitary pulmonary nodule in breast cancer patients. Eur J Surg Oncol 2007;33:546-50.
  25. Welter S, Jacobs J, Krbek T, Tötsch M, Stamatis G. Pulmonary metastases of breast cancer. When is resection indicated? Eur J Cardiothorac Surg 2008;34:1228-34.
  26. Borner M, Bacchi M, Goldhirsch A, Greiner R, Harder F, Castiglione M, et al. First isolated locoregional recurrence following mastectomy for breast cancer: Results of a phase III multicenter study comparing systemic treatment with observation after excision and radiation. Swiss Group for Clinical Cancer Research. J Clin Oncol 1994;12:2071-7.
  27. Aebi S, Gelber S, Anderson SJ, Láng I, Robidoux A, Martín M, et al. Chemotherapy for isolated locoregional recurrence of breast cancer (CALOR): A randomised trial. Lancet Oncol 2014;15:156-63.