430
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Breast Cancer: An Overview of Current Therapeutic Strategies, Challenge, and Perspectives

ORCID Icon &
Pages 721-730 | Received 26 Jul 2023, Accepted 11 Oct 2023, Published online: 20 Oct 2023

Abstract

Breast cancer is the most commonly diagnosed cancer and the leading cause of death among female patients, which seriously threatens the health of women in the whole world. The treatments of breast cancer require the cooperation of a multidisciplinary setting and taking tumor load and molecular makers into account. For early breast cancer, breast-conserving surgery with radiotherapy or mastectomy alone remains the standard management, and the administration of adjuvant systemic therapy is decided by the status of lymph nodes, hormone receptors, and human epidermal growth factor receptor-2. For metastatic breast cancer, the goal of treatments is to prolong survival and maintain quality of life. This review will present the current advances and controversies of surgery, chemotherapy, radiotherapy, endocrine therapy, targeted therapy, immunotherapy, gene therapy, and other innovative treatment strategies in early-stage and metastatic breast cancer.

Introduction

Breast cancer is the most commonly diagnosed cancer among female patients and is the leading cause of cancer-related death.Citation1 There were 300,590 new cases and 43,700 deaths of invasive breast cancer in the United States based on the 2023 prediction, accounting for approximately 30% of female cancers.Citation1 The treatments of breast cancer include surgery, chemotherapy, radiotherapy (RT), endocrine therapy, targeted therapy, and immunotherapy, and the therapeutic schedules require the cooperation of multiple subspecialties. For non-metastatic breast cancer, surgery-based treatment is the standard management, and chemotherapy-based preoperative systemic therapy can reduce tumor volume of the breast, making breast conservation possible, and decreasing the need for axillary lymph node dissection (ALND).Citation2 Systemic treatment remains the preferred option for metastatic breast cancer, and surgery is only used for palliative therapy in selected metastatic patients.Citation3 The advances in endocrine therapy, targeted therapy, and immunotherapy provide additional treatments for patients with metastatic and non-metastatic breast cancer. Some innovative therapies are also being investigated, such as gene therapy, vaccines, adoptive cell therapies, including T cell receptor therapy and chimeric antigen receptor T (CAR-T) therapy, and achieved promising results. This review aims to summarize the current status and controversies of surgery, chemotherapy, RT, endocrine therapy, targeted therapy, immunotherapy, gene therapy, and other innovative therapies in breast cancer, and provides better management for oncologists.

Surgery

Breast-conserving surgery (BCS) and mastectomy with or without immediate reconstruction are both well-established local managements for early invasive breast cancer. The widespread use of systematic treatments in past decades led to the reduction of locoregional recurrence rates (LRR) and distant metastasis rates, and the 10-year LRR of BCS followed by RT was 2–3% for estrogen receptor (ER) positive and human epidermal growth factor receptor-2 (HER-2) positive breast cancer and 5% for triple-negative breast cancer (TNBC), which was similar to that after mastectomy in early breast cancer.Citation4,Citation5 In addition, patients with BCS+RT had better cosmetic effects and life satisfaction compared with mastectomy.Citation6 Therefore, BCS following RT is the intended surgical standard of care for most breast cancers. However, the selection of BCS should still be cautious for patients with diffuse suspicious micro-calcifications, multicentric cancer, unable to obtain negative margins, and having contraindications to RT.Citation7 Younger age, lobular carcinoma, and aggressive subtypes, such as triple-negative and HER2 positive diseases are not contraindications for BCS. For patients with large tumors, neoadjuvant chemotherapy (NAC) can be chosen to downstage the tumor for BCS.

The management of axillary lymph nodes (ALNs) is decided by the status of ALNs at diagnosis and the administration of neoadjuvant systemic therapy. ALND remains the standard in patients with clinically proven axillary involvement at initial diagnosis.Citation8 For patients with clinically node-negative (cN0) breast cancer, the management of axillary is controversial. There seemed to be comparable recurrence and survival outcomes between ALND and sentinel lymph node (SLN) biopsy in the era of contemporary systemic treatments.Citation9 In addition, several prospective trials demonstrated that there was no significance of recurrence and survival between SLN biopsy alone and SLN biopsy plus RT in patients with cN0 and one to two SLN involvement.Citation10–12 Therefore, SLN biopsy might be sufficient for most cN0 patients, and additional axillary radiation only for selected patients, such as patients with three SLN involvement. Further studies are needed to explore the value of ALND, SLN biopsy, and RT in patients with cN0 disease.

Surgery also plays an important role in the management of local-regional recurrent breast cancer. Total mastectomy remains the standard of care for recurrent patients initially receiving BCS, and salvage mastectomy ± ALND could achieve 85–95% loco-regional control in this disease.Citation8,Citation13 Patients suffering chest-wall recurrence after initial mastectomy had a higher risk of metastasis than those initially treated with BCS.Citation14 In addition, previous studies showed that limited resection was related to a higher second local recurrence of 60–70%; therefore, the routinely recommended management for patients initially treated with mastectomy is wide resection of the recurrent lesions when possible.Citation15,Citation16

Chemotherapy

The modalities of chemotherapy in breast cancer include NAC, adjuvant chemotherapy (AC), and salvage chemotherapy. The administration of chemotherapy could reduce the risk of recurrence by approximately 30% in early breast cancer.Citation17 NAC could downstage the breast and axilla for receiving breast conservation in operable breast cancer, convert inoperable breast cancer to resectable, and eliminate micrometastatic lesions. NAC is recommended for patients with a large tumor, multiple ALNs involvement, and aggressive subtypes, especially for triple-negative and HER2-positive breast cancer.Citation18 The application of NAC in aggressive breast cancer subtypes could evaluate the response to treatment, predict cancer prognosis, and guide subsequent treatment decision-making. Previous studies showed that NAC followed by surgery had similar LRR and survival outcomes compared with surgery followed by chemotherapy, but patients treated with NAC decreased 17% of mastectomy rates.Citation19,Citation20 NAC or AC should be administrated in high-risk patients, such with large tumor size, nodal involvement, low expression of hormone receptor (HR), younger age, and lymphovascular invasion.Citation8 Sandwich chemotherapy should be avoided outside of clinical trials.Citation21 In addition, multigene assays and molecular types could be used to stratify and identify patients who benefit from chemotherapy, especially for node-negative ER-positive, triple-negative, or HER2-positive diseases.Citation22–24

The current optimal chemotherapy regimen is taxane with or without anthracycline, given in sequence or combination, both in NAC and AC settings.Citation8 The application of anthracyclines remains controversial, but it seems to be essential in high-risk patients, such as triple-negative and HER-2 positive subtypes.Citation25,Citation26 For patients with TNBC, the addition of platinum to standard regimens (anthracycline and taxane-based regimen) achieved an absolute improvement of pathological complete response (pCR) (15%); however, long-term survival outcomes are less convincing due to the small sample size and short follow-up time.Citation27,Citation28 Another Phase 3 randomized clinical trial including 647 patients explored the value of docetaxel and carboplatin regimens in place of standard regimens in patients with TNBC, and docetaxel and carboplatin showed improved disease-free survival (DFS) and relapse-free survival, but comparable overall survival (OS) compared with standard regimens.Citation29 Therefore, the docetaxel and carboplatin regimen can be used as an alternative to the conventional regimen in the treatment of TNBC. In addition, another Phase III study included 443 patents with early TNBC to assess the effect of low-dose and high-frequency capecitabine maintenance, and result showed that patients receiving capecitabine maintenance had higher 5-year distant disease-free survival (85.8% vs 75.8%, p = 0.02); therefore, low-dose and high-frequency capecitabine maintenance could be used as consolidation therapy in patients receiving standard adjuvant therapy.Citation30,Citation31

AC is commonly recommended for 3–4 weeks after surgery.Citation32 A population-based study explored the effect of the wider time window, and the authors found that delays beyond 91 days from surgery to the start of AC were related to worse survival, especially in TNBC.Citation33 Therefore, the administration of AC within 3 months after breast surgery is acceptable. With regard to optimal chemotherapy interval, a dose-dense regimen (every 14 days) had better DFS and OS than the conventional interval (every 21 days) in early breast cancer.Citation34

Radiotherapy

Two large-scale randomized trials evaluating the effect of omitting RT in low-risk patients receiving BCS achieved negative results, thus adjuvant RT remains the standard of care for patients receiving BCS.Citation35,Citation36 Whole breast irradiation (WBI) is a convenient option used for adjuvant RT following BCS. Hypofractionated WBI appeared to have comparable local recurrence, survival outcomes, and toxicity profiles compared with WBI.Citation37,Citation38 Therefore, hypofractionated WBI is also supported for early breast cancer not requiring nodal therapy.Citation37,Citation38 In addition, partial breast irradiation (PBI) demonstrated similar local recurrence, but improved cosmetic results and reduced toxicity compared with WBI, thus PBI seemed to be an acceptable alternative for appropriately selected low-risk patients.Citation39,Citation40 Accelerated partial-breast irradiation (APBI) is also a treatment option for selected early-stage breast cancer patients receiving BCS with reduced recurrence rates (HR = 4.54, 95% CI: 1.78–11.61, p = 0.002) compared with WBI.Citation41 Brachytherapy also showed a similar breast cancer recurrence rate compared with external radiotherapy (5-year event rate: 4·4%) in the radiation of breast cancer, yet the application of brachytherapy should be cautious due to the lack of sufficient evident data.Citation42

For patients receiving mastectomy, whether to irradiate is decided by the number of involved ALNs. Postmastectomy radiation therapy (PMRT) is conventionally used in patients with four or more positive ALNs;Citation8 however, the administration of PMRT in patients with one to three involved ALNs remains unclear. Current clinical guidelines strongly recommend PMRT for this patient subset, which is based on the result from the meta-analysis published in Lancet 2014 that PMRT had significantly decreased the LRR rate and cancer-related death.Citation8,Citation43 However, most of the trials enrolled in this meta-analysis were completed before the 1980s, when the RT technique and chemotherapy regimens were much more backward than what it is now, and the LRR rate without PMRT was approximately 30% at that time.Citation43 Several recent studies showed that there seemed no incremental survival benefits in patients with one to three involved ALNs in the era of contemporary systemic treatment.Citation44,Citation45 Therefore, it seems to be essential to select high-risk patients for receiving PMRT, such as younger age, a higher burden of breast and axilla, and biological characteristics. Recent studies demonstrated that the 8th American Joint Committee on Cancer (AJCC) pathological prognostic staging integrating molecular markers could guide the RT administration in patients with N1 breast cancer.Citation46,Citation47 Further studies are needed to explore the value of PMRT in patients with one to three involved ALNs.

For patients with positive regional lymph nodes, there is evidence that chest wall and infra-/supraclavicular regions radiation were beneficial. The MA.20 trial included 1832 patients with LN positive, and randomized to receive breast irradiation with or without full regional nodal irradiation [including internal mammary nodes (IMNs)].Citation48 Ten-year follow-up showed that additional regional nodal irradiation had improved 24% DFS.Citation48 Chest wall and infra-/supraclavicular regions are commonly irradiation areas; however, the value of IMNs radiation remains a debate. Several studies demonstrated improved OS and decreased breast cancer mortality in patients receiving regional nodal irradiation including IMNs, but higher heart and lung toxicities, lymphedema, and non-breast cancer mortality risk were observed.Citation49,Citation50 Another trial evaluated the value of additional IMN irradiation in patients with positive ALNs or central lesions without positive ALNs, and the result showed that there was no survival benefit in any of the groups (10-year OS: 62.6% vs 59.3%, P > 0.5). Therefore, further studies should be conducted to explore the effect of IMN irradiation.Citation51

Endocrine Therapy

Endocrine therapy is considered standard as adjuvant therapy for patients with HR-positive (ER or PR staining ≥1%) over the course of 5–10 years. The sensitivity of endocrine therapy is directly associated with the expression of hormone receptors.Citation52 In premenopausal patients, tamoxifen 20 mg per day for 5 years could reduce about 50% recurrence risk in the first 4 years, and over 30% during 5–9 years, and a longer duration of tamoxifen resulted in further reduction of recurrence and breast cancer mortality.Citation53 Another study from the ATLAS trial demonstrated that the administration of 10-year tamoxifen could reduce breast cancer death by 2% (9.6% vs 11.6%) compared with 5-year tamoxifen.Citation54 Therefore, the duration of tamoxifen should be continued for 10 years. For patients with a high risk of relapses, such as those aged ≤35 years or after chemotherapy, ovarian suppression drugs combined with an aromatase inhibitor (AI) or tamoxifen had improved DFS, but higher toxicity compared with tamoxifen alone.Citation55,Citation56

In postmenopausal patients, tamoxifen or AI monotherapy 5 years or in sequence are both alternative treatment strategies. AI monotherapy 5 years had reduced breast cancer mortality than tamoxifen 5 years, thus AI is preferred as adjuvant therapy, especially in high-risk patients and patients with lobular histology.Citation57,Citation58 However, patients receiving AI therapy had higher rates of bone-related adverse events, such as fractures and osteoporosis; therefore, tamoxifen can be used as an alternative for patients with serious AI-related adverse events. The duration of tamoxifen and AI in postmenopausal patients should be further assessed for the balance of risks and benefits, and multigene assays might be useful in predicting the appropriate duration.Citation59 In addition, the combination of CDK inhibitors and AI could significantly improve DFS in metastatic breast cancer compared with AI alone; therefore, CDK inhibitors plus AI could be an alternative strategy in endocrine-resistant metastatic breast cancer.Citation60 Detailed information is presented in .

Table 1 Representative Studies of Endocrine Therapy for Breast Cancer

The value of neoadjuvant endocrine therapy in early ER-positive breast cancer was unclear. Early studies of neoadjuvant endocrine therapy were focused on elderly postmenopausal patients with locally advanced breast cancer, or unable to receive chemotherapy. The response rate of neoadjuvant tamoxifen was about 50%.Citation63,Citation64 A recent study showed that neoadjuvant endocrine therapy combined with chemotherapy could increase BCS rates in postmenopausal patients with luminal breast cancer.Citation61 Moreover, neoadjuvant anastrozole seemed to have a similar anti-cancer effect compared with tamoxifen.Citation62 AI is more commonly used as neoadjuvant therapy than tamoxifen for better efficacy. Currently, there is no evidence for the optimum duration of neoadjuvant endocrine therapy lacking supporting data. In addition, other ongoing trials use the level of Ki67 after neoadjuvant endocrine therapy to guide the administration of AC.

Targeted Therapy

The appearance of anti-HER2 targeted therapy greatly changed the treatment paradigm and prognosis of HER2-positive breast cancer. Trastuzumab, the first anti-HER2 targeted drug, has been widely used in HER2-positive diseases. American Society of Clinical Oncology (ASCO) Annual Meeting in 2005 firstly reported the benefit of trastuzumab combined with anthracycline/taxane-based adjuvant therapy in HER2-positive breast cancer. A later study showed that adjuvant trastuzumab combined with paclitaxel had low local-regional and distant recurrences and reduced toxicity in HER-2 positive breast cancer patients with tumors ≤2 cm and negative nodes.Citation65 In addition, trastuzumab combined with other chemotherapy regimens (adriamycin/ cyclophosphamide-paclitaxel, docetaxel, and carboplatin) achieved absolute OS advantage.Citation66 Based on the above studies, trastuzumab (1 year) incorporating chemotherapy (adriamycin/cyclophosphamide-paclitaxel, docetaxel, and carboplatin) as neoadjuvant and adjuvant regimens is recommended for patients with HER2-positive disease.Citation8

Other anti-HER2 drugs, such as pertuzumab and lapatinib, were also assessed in HER-2-positive breast cancer. Recent prospective data showed that additional pertuzumab to trastuzumab plus docetaxel in neoadjuvant therapy could significantly improve pCR compared with trastuzumab plus docetaxel (45·8% vs 29.0%); therefore, neoadjuvant dual-HER2 agents (pertuzumab/trastuzumab) are also an alternative for stage II–III HER2 positive breast cancer.Citation67 Lapatinib seemed to have higher pCR in the neoadjuvant setting, and a recent phase III study showed that paclitaxel combined with trastuzumab plus lapatinib had better RFS and OS compared with paclitaxel plus lapatinib; thus, chemotherapy plus dual HER2-targeting drugs was still a promising treatment in HER2-positive breast cancer.Citation68

For metastatic HER2-positive breast cancer, anti-HER2 therapy should be used as early as possible, docetaxel plus trastuzumab and pertuzumab were recommended as first-line standards based on the CLEOPATRA trial.Citation69 Trastuzumab emtansine (TDM-1) had a significantly prolonged PFS and OS than lapatinib plus capecitabine in metastatic HER2-positive breast cancer patients previously treated with trastuzumab from the EMILIA trial.Citation70 Therefore, lapatinib has been approved for the second-line treatment of metastatic HER2-positive breast cancer. Neratinib is another small molecule of HER1/HER2 inhibitors. The addition of neratinib to capecitabine showed improved PFS and lower central nervous system disease compared with lapatinib plus capecitabine in the phase III NALA trial.Citation71 Another Phase II trial evaluated the value of single-agent neratinib in metastatic HER2-positive breast cancer, but only 8% response rates were observed; thus, the value of neratinib in metastatic breast cancer should be further assessed.Citation72 Despite the advances in targeted therapy in HER2-positive breast cancer, the resistance to anti-HER2 drugs is still a serious problem and exploring the way to relieve targeted therapy resistance is needed.

Immunotherapy

Breast cancer has impaired activated T cell killing of tumor cells due to the presence of inhibitory factors such as interaction between the PD-1, TIM-3, LAG3, TIGIT, CTLA4 and their ligands on the cancer cells promotes T-cell exhaustion and prevents responsiveness to therapy. Therefore, the use of immune checkpoint blockade as an anti-tumor therapy has demonstrated modest single agent activity in advanced breast cancer.Citation73,Citation74

The efficacy of single-agent immune checkpoint inhibitors (ICIs) in metastatic TNBC is low.Citation75–77 KEYNOTE-012 trial evaluated the value of pembrolizumab in pre-treated PD-L1 positive metastatic TNBC, and an overall response rate (ORR) of 18.5% was observed.Citation75 KEYNOTE-086 and JAVELIN trials showed an ORR of 21.4% (pembrolizumab) and 5.2% (avelumab), respectively.Citation76,Citation77 The combination of chemotherapy with ICIs demonstrated better results than ICI monotherapy, with an ORR of 26.4–39.4% in 0–2 prior lines of treatment for metastatic TNBC.Citation78,Citation79 IMpassion130, a phase III large-scale randomized trial, included 902 previously untreated, locally advanced, or metastatic TNBC patients, and randomized to receive atezolizumab plus nab-paclitaxel or nab-paclitaxel alone.Citation80 The result showed that patients treated with atezolizumab plus nab-paclitaxel had significantly improved median OS compared with nab-paclitaxel alone.Citation80 However, another study from the IMpassion131 trial achieved a negative result that atezolizumab combined with paclitaxel could not improve survival compared with paclitaxel.Citation81 In addition, pembrolizumab plus chemotherapy (nab-paclitaxel; paclitaxel; or gemcitabine plus carboplatin) had increased PFS than chemotherapy alone in untreated locally recurrent or metastatic TNBC.Citation82 Therefore, atezolizumab and pembrolizumab are approved by the United States Food and Drug Administration (FDA) for the first-line treatment of PD-L1 positive metastatic TNBC. In addition, in patients with metastatic HER2-positive breast cancer, pembrolizumab+trastuzumab showed a 15% response rates in PD-L1-positive tumors; however, atezolizumab combined with T-DM1 did not improve PFS but increased toxicity.Citation83,Citation84

The value of ICIs plus chemotherapy was also explored in early-stage breast cancer, and preliminary success was observed.Citation85,Citation86 The addition of pembrolizumab to chemotherapy (paclitaxel, doxorubicin, cyclophosphamide) in the neoadjuvant setting of stage II–III breast cancer had higher pCR rates in HER2-negative (44% vs 17%), HR-positive/HER2-negative (30% vs 13%), and triple-negative subtypes (60% vs 22%) than chemotherapy alone.Citation85 KEYNOTE-522 demonstrated that four cycles of pembrolizumab plus paclitaxel/ carboplatin, then four cycles of pembrolizumab plus anthracycline/cyclophosphamide showed improved pCR rates (64.8% vs 51.2%) and event-free survival (91.3% vs 85.3%) compared with neoadjuvant chemotherapy alone in TNBC.Citation86 In addition, neoadjuvant durvalumab plus nab-paclitaxel/epirubicin/cyclophosphamide had an increased pCR rate, especially in patients with durvalumab alone before chemotherapy.Citation87 In Impassion 031 study, atezolizumab combined with nab-paclitaxel+anthracycline also had increased pCR rate (41% vs 58%, p = 0.0044) in TNBC patients.Citation85 However, fewer results were available in luminal and HER2-positive subtypes. A Phase 2 study exploring the effect of pembrolizumab plus neoadjuvant paclitaxel+doxorubicin+cyclophosphamide in HER2-negative patients, and results showed that patients receiving pembrolizumab undergone higher pCR rate (30% vs 13%) in luminal patients.Citation88 Although promising results were observed in neoadjuvant therapy of ICIs plus chemotherapy, longer follow-up data are needed to confirm the long-term efficacy.

Gene Therapy

Gene therapy is also a promising approach in the treatment of cancers, which defined as sending genetic material through a vector into target cells to edit the gene and change the expression of a gene’s product, and achieving the goal of treating cancers.Citation89 Gene therapy strategies include gene editing, targeting transcription factors, microRNA, and breast cancer cells, DNA or RNA vaccination, and so on. A Phase I clinical trials tested the safety and efficacy of genetic prodrug activation therapy targeted the human HER-2 gene promoter. The study included 12 breast cancer patients, and result showed that the approach was safe and targeted gene expression was detected in up to 90% of the patients.Citation90 Another phase 2 trial includes 28 patients with metastatic TNBC to explore the value of in situ virus gene therapy (ADV/HSV-tk) plus stereotactic body radiotherapy and pembrolizumab, and the result demonstrated that clinical benefit rate was 21.4%, and patients with clinical benefit had durable responses, with improved median duration on treatment (9.6 months) and OS (14.7 months).Citation91 Use of microRNA in anti-cancer therapy also showed promising results in inhibiting breast cancer cell proliferation and development. MRX34, to our knowledge, is one of the first miRNA replacement agents (miR-34a), and now in entering clinical trials. It is believed that it will play a vital role in the treatment of breast cancer in the future. Currently, few studies on gene therapy were published, but a lot of strategies have entered clinical trials in breast cancer. We summarized several clinical studies on breast cancer gene therapy in .

Table 2 Ongoing Trials of Gene Therapy, Vaccine, and CAR-T Therapy of Breast Cancer

Conclusion and Future Perspectives

The value of local and systemic therapies in breast cancer has been well established. For early breast cancer, surgery-based local and systemic treatments are the standard of care. For metastatic breast cancer, chemotherapy-based systemic treatments remain the preferred option, and surgery is only used for palliative therapy in selected patients. However, survival benefits of traditional treatment strategies were limited. The emergence of targeted therapy and immunotherapy further changed the treatment pattern of early and metastatic breast cancer. Atezolizumab or pembrolizumab combined with chemotherapy are approved by FDA for the first-line treatment of PD-L1 positive metastatic TNBC. Neoadjuvant and adjuvant pembrolizumab is approved for early TNBC. Numerous novel ICIs and new ICIs-based combination therapies have entered into clinical trails.

In addition, some innovative therapies are being investigated in breast cancer, such as gene therapy, breast cancer vaccines, adoptive cell therapies, including T cell receptor therapy and CAR-T therapy, and so on. Several phase I/II clinical trails have demonstrated an preliminary improved outcome, and most of them are still ongoing. We summarize some ongoing clinical studies in . We believe these new therapeutic strategies will gradually be integrated into clinical treatments.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48. doi:10.3322/caac.21763
  • Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019;30(8):1194–1220. doi:10.1093/annonc/mdz173
  • Senkus E, Łacko A. Over-treatment in metastatic breast cancer. Breast. 2017;31:309–317. doi:10.1016/j.breast.2016.06.024
  • Zumsteg ZS, Morrow M, Arnold B, et al. Breast-conserving therapy achieves locoregional outcomes comparable to mastectomy in women with T1-2N0 triple-negative breast cancer. Ann Surg Oncol. 2013;20(11):3469–3476. doi:10.1245/s10434-013-3011-9
  • van Maaren MC, de Munck L, Strobbe LJA, et al. Ten-year recurrence rates for breast cancer subtypes in the Netherlands: a large population-based study. Int J Cancer. 2019;144(2):263–272. doi:10.1002/ijc.31914
  • Cipora E, Konieczny M, Karwat ID, et al. Surgical method of treatment and level of satisfaction with life among women diagnosed with breast cancer, according to time elapsed since performance of surgery. Ann Agric Environ Med. 2018;25(3):453–459. doi:10.26444/aaem/91586
  • Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321(3):288–300. doi:10.1001/jama.2018.19323
  • Breast cancer. National Comprehensive Cancer Network: National Clinical Practice Guidelines in Oncology. Version 2; 2022. Available from: https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf. Accessed October 13, 2023.
  • Krag DN, Anderson SJ, Julian TB, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. 2010;11(10):927–933.
  • Giuliano AE, Ballman KV, McCall L, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: the ACOSOG Z0011 (Alliance) Randomized Clinical Trial. JAMA. 2017;318(10):918–926. doi:10.1001/jama.2017.11470
  • Donker M, van Tienhoven G, Straver ME, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981–22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet Oncol. 2014;15(12):1303–1310. doi:10.1016/S1470-2045(14)70460-7
  • Sávolt Á, Péley G, Polgár C, et al. Eight-year follow up result of the OTOASOR trial: the Optimal Treatment of the Axilla - Surgery Or Radiotherapy after positive sentinel lymph node biopsy in early-stage breast cancer: a randomized, single centre, Phase III, non-inferiority trial. Eur J Surg Oncol. 2017;43(4):672–679. doi:10.1016/j.ejso.2016.12.011
  • Wilson RE. Surgical management of locally advanced and recurrent breast cancer. Cancer. 1984;53(3 Suppl):752–757.
  • Early Breast Cancer Trialists’ Collaborative Group. Favourable and unfavourable effects on long-term survival of radiotherapy for early breast cancer: an overview of the randomised trials. Lancet. 2000;355(9217):1757–1770. doi:10.1016/S0140-6736(00)02263-7
  • Donegan WL, Perez-Mesa CM, Watson FR. A biostatistical study of locally recurrent breast carcinoma. Surg Gynecol Obstet. 1966;122(3):529–540.
  • Halverson KJ, Perez CA, Kuske RR, et al. Isolated local-regional recurrence of breast cancer following mastectomy: radiotherapeutic management. Int J Radiat Oncol Biol Phys. 1990;19(4):851–858. doi:10.1016/0360-3016(90)90004-4
  • Mano MS, Awada A. Primary chemotherapy for breast cancer: the evidence and the future. Ann Oncol. 2004;15(8):1161–1171. doi:10.1093/annonc/mdh302
  • Moo TA, Sanford R, Dang C, et al. Overview of breast cancer therapy. PET Clin. 2018;13(3):339–354. doi:10.1016/j.cpet.2018.02.006
  • Rastogi P, Anderson SJ, Bear HD, et al. Preoperative chemotherapy: updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol. 2008;26(5):778–785. doi:10.1200/JCO.2007.15.0235
  • Mieog JS, van der Hage JA, van de Velde CJ. Neoadjuvant chemotherapy for operable breast cancer. Br J Surg. 2007;94(10):1189–1200. doi:10.1002/bjs.5894
  • Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389(10074):1134–1150. doi:10.1016/S0140-6736(16)31891-8
  • Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med. 2018;379(2):111–121. doi:10.1056/NEJMoa1804710
  • Andre F, Ismaila N, Henry NL, et al. Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: ASCO clinical practice guideline update-integration of results from TAILORx. J Clin Oncol. 2019;37(22):1956–1964. doi:10.1200/JCO.19.00945
  • Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–1717. doi:10.1016/S0140-6736(05)66544-0
  • Blum JL, Flynn PJ, Yothers G, et al. Anthracyclines in Early Breast Cancer: the ABC Trials-USOR 06-090, NSABP B-46-I/USOR 07132, and NSABP B-49 (NRG Oncology). J Clin Oncol. 2017;35(23):2647–2655. doi:10.1200/JCO.2016.71.4147
  • Nitz U, Gluz O, Clemens M, et al. West German Study Plan B Trial: adjuvant four cycles of epirubicin and cyclophosphamide plus docetaxel versus six cycles of docetaxel and cyclophosphamide in HER2-negative early breast cancer. J Clin Oncol. 2019;37(10):799–808. doi:10.1200/JCO.18.00028
  • Poggio F, Bruzzone M, Ceppi M, et al. Platinum-based neoadjuvant chemotherapy in triple-negative breast cancer: a systematic review and meta-analysis. Ann Oncol. 2018;29(7):1497–1508. doi:10.1093/annonc/mdy127
  • von Minckwitz G, Schneeweiss A, Loibl S, et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol. 2014;15(7):747–756. doi:10.1016/S1470-2045(14)70160-3
  • Yu KD, Ye FG, He M, et al. Effect of adjuvant paclitaxel and carboplatin on survival in women with triple-negative breast cancer: a Phase 3 randomized clinical trial. JAMA Oncol. 2020;6(9):1390–1396. doi:10.1001/jamaoncol.2020.2965
  • Wang X, Wang SS, Huang H, et al. Effect of capecitabine maintenance therapy using lower dosage and higher frequency vs observation on disease-free survival among patients with early-stage triple-negative breast cancer who had received standard treatment: the SYSUCC-001 randomized clinical trial. JAMA. 2021;325(1):50–58. doi:10.1001/jama.2020.23370
  • Parsons HA, Burstein HJ. Adjuvant capecitabine in triple-negative breast cancer: new strategies for tailoring treatment recommendations. JAMA. 2021;325(1):36–38. doi:10.1001/jama.2020.23371
  • Downing A, Twelves C, Forman D, et al. Time to begin adjuvant chemotherapy and survival in breast cancer patients: a retrospective observational study using latent class analysis. Breast J. 2014;20(1):29–36. doi:10.1111/tbj.12209
  • Chavez-MacGregor M, Clarke CA, Lichtensztajn DY, et al. Delayed initiation of adjuvant chemotherapy among patients with breast cancer. JAMA Oncol. 2016;2(3):322–329. doi:10.1001/jamaoncol.2015.3856
  • Citron ML, Berry DA, Cirrincione C, et al. Randomized trial of dose-dense versus conventionally scheduled and sequential versus concurrent combination chemotherapy as postoperative adjuvant treatment of node-positive primary breast cancer: first report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741. J Clin Oncol. 2003;21(8):1431–1439. doi:10.1200/JCO.2003.09.081
  • Hughes KS, Schnaper LA, Bellon JR, et al. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: long-term follow-up of CALGB 9343. J Clin Oncol. 2013;31(19):2382–2387. doi:10.1200/JCO.2012.45.2615
  • Kunkler IH, Williams LJ, Jack WJ, et al. Breast-conserving surgery with or without irradiation in women aged 65 years or older with early breast cancer (PRIME II): a randomised controlled trial. Lancet Oncol. 2015;16(3):266–273. doi:10.1016/S1470-2045(14)71221-5
  • Whelan TJ, Pignol JP, Levine MN, et al. Long-term results of hypofractionated radiation therapy for breast cancer. N Engl J Med. 2010;362(6):513–520. doi:10.1056/NEJMoa0906260
  • Haviland JS, Owen JR, Dewar JA, et al. The UK Standardisation of Breast Radiotherapy (START) trials of radiotherapy hypofractionation for treatment of early breast cancer: 10-year follow-up results of two randomised controlled trials. Lancet Oncol. 2013;14(11):1086–1094. doi:10.1016/S1470-2045(13)70386-3
  • Strnad V, Ott OJ, Hildebrandt G, et al. 5-year results of accelerated partial breast irradiation using sole interstitial multicatheter brachytherapy versus whole-breast irradiation with boost after breast-conserving surgery for low-risk invasive and in-situ carcinoma of the female breast: a randomised, phase 3, non-inferiority trial. Lancet. 2016;387(10015):229–238. doi:10.1016/S0140-6736(15)00471-7
  • Coles CE, Griffin CL, Kirby AM, et al. Partial-breast radiotherapy after breast conservation surgery for patients with early breast cancer (UK IMPORT LOW trial): 5-year results from a multicentre, randomised, controlled, phase 3, non-inferiority trial. Lancet. 2017;390(10099):1048–1060. doi:10.1016/S0140-6736(17)31145-5
  • Marta GN, Macedo CR, Carvalho Hde A, et al. Accelerated partial irradiation for breast cancer: systematic review and meta-analysis of 8653 women in eight randomized trials. Radiother Oncol. 2015;114(1):42–49. doi:10.1016/j.radonc.2014.11.014
  • Veronesi U, Orecchia R, Maisonneuve P, et al. Intraoperative radiotherapy versus external radiotherapy for early breast cancer (ELIOT): a randomised controlled equivalence trial. Lancet Oncol. 2013;14(13):1269–1277. doi:10.1016/S1470-2045(13)70497-2
  • Kwong DL, McGale P, Taylor C, et al; Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Effect of radiotherapy after mastectomy and axillary surgery on 10-year recurrence and 20-year breast cancer mortality: meta-analysis of individual patient data for 8135 women in 22 randomised trials. Lancet. 2014;383(9935):2127–2135. doi:10.1016/S0140-6736(14)60488-8
  • McBride A, Allen P, Woodward W, et al. Locoregional recurrence risk for patients with T1,2 breast cancer with 1–3 positive lymph nodes treated with mastectomy and systemic treatment. Int J Radiat Oncol Biol Phys. 2014;89(2):392–398. doi:10.1016/j.ijrobp.2014.02.013
  • Muhsen S, Moo TA, Patil S, et al. Most breast cancer patients with T1–2 tumors and one to three positive lymph nodes do not need postmastectomy radiotherapy. Ann Surg Oncol. 2018;25(7):1912–1920. doi:10.1245/s10434-018-6422-9
  • Wu SG, Wang J, Lian CL, et al. Evaluation of the 8th edition of the American joint committee on cancer’s pathological staging system in prognosis assessment and treatment decision making for stage T1-2N1 breast cancer after mastectomy. Breast. 2020;51:2–10. doi:10.1016/j.breast.2020.02.012
  • Wang S, Wen G, Tang Y, et al. Effectiveness of the AJCC 8th edition staging system for selecting patients with T1-2N1 breast cancer for post-mastectomy radiotherapy: a joint analysis of 1986 patients from two institutions. BMC Cancer. 2020;20(1):792. doi:10.1186/s12885-020-07267-5
  • Whelan TJ, Olivotto IA, Parulekar WR, et al. Regional nodal irradiation in early-stage breast cancer. N Engl J Med. 2015;373(4):307–316. doi:10.1056/NEJMoa1415340
  • Poortmans PM, Collette S, Kirkove C, et al. Internal mammary and medial supra-clavicular irradiation in breast cancer. N Engl J Med. 2015;373(4):317–327. doi:10.1056/NEJMoa1415369
  • Thorsen LBJ, Offersen BV, Dano H, et al. DBCG-IMN: a population-based cohort study on the effect of internal mammary node irradiation in early node-positive breast cancer. J Clin Oncol. 2016;34(4):314–320. doi:10.1200/JCO.2015.63.6456
  • Hennequin C, Bossard N, Servagi-Vernat S, et al. Ten-year survival results of a randomized trial of irradiation of internal mammary nodes after mastectomy. Int J Radiat Oncol Biol Phys. 2013;86(5):860–866. doi:10.1016/j.ijrobp.2013.03.021
  • Du L, Yau C, Brown-Swigart L, et al. Predicted sensitivity to endocrine therapy for stage II-III hormone receptor-positive and HER2-negative (HR+/HER2-) breast cancer before chemo-endocrine therapy. Ann Oncol. 2021;32(5):642–651. doi:10.1016/j.annonc.2021.02.011
  • Davies C, Godwin J. Early Breast Cancer Trialists’ Collaborative Group. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet. 2011;378(9793):771–784. doi:10.1016/S0140-6736(11)60993-8
  • Davies C, Pan H, Godwin J, et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: a TLAS, a randomised trial. Lancet. 2013;381(9869):805–816. doi:10.1016/S0140-6736(12)61963-1
  • Pagani O, Regan MM, Walley BA, et al. Adjuvant exemestane with ovarian suppression in premenopausal breast cancer. N Engl J Med. 2014;371:107–118. doi:10.1056/NEJMoa1404037
  • Francis PA, Regan MM, Fleming GF, et al. Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med. 2015;372(5):436–446. doi:10.1056/NEJMoa1412379
  • Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials. Lancet. 2015;386(10001):1341–1352. doi:10.1016/S0140-6736(15)61074-1
  • Dowsett M, Cuzick J, Ingle J, et al. Meta-analysis of breast cancer outcomes in adjuvant trials of aromatase inhibitors versus tamoxifen. J Clin Oncol. 2010;28(3):509–518. doi:10.1200/JCO.2009.23.1274
  • Knauer M, Filipits M, Dubsky P. Late recurrences in early breast cancer: for whom and how long is endocrine therapy beneficial? Breast Care. 2014;9(2):97–100. doi:10.1159/000362482
  • Gao JJ, Cheng J, Bloomquist E, et al. CDK4/6 inhibitor treatment for patients with hormone receptor-positive, HER2-negative, advanced or metastatic breast cancer: a US Food and Drug Administration pooled analysis. Lancet Oncol. 2020;21(2):250–260. doi:10.1016/S1470-2045(19)30804-6
  • Alba E, Calvo L, Albanell J, et al. Chemotherapy (CT) and hormonotherapy (HT) as neoadjuvant treatment in luminal breast cancer patients: results from the GEICAM/2006-03, a multicenter, randomized, phase-II study. Ann Oncol. 2012;23(12):3069–3074. doi:10.1093/annonc/mds132
  • Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the immediate preoperative Anastrozole, Tamoxifen, or combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol. 2005;23(22):5108–5116. doi:10.1200/JCO.2005.04.005
  • Horobin JM, Preece PE, Dewar JA, et al. Long-term follow-up of elderly patients with locoregional breast cancer treated with tamoxifen only. Br J Surg. 1991;78(2):213–217. doi:10.1002/bjs.1800780228
  • Gaskell DJ, Hawkins RA, de Carteret S, et al. Indications for primary tamoxifen therapy in elderly women with breast cancer. Br J Surg. 1992;79(12):1317–1320. doi:10.1002/bjs.1800791225
  • Tolaney SM, Barry WT, Guo H, et al. Seven-year follow-up of adjuvant paclitaxel and trastuzumab (APT trial) for node-negative, HER2+ breast cancer. J Clin Oncol. 2017;35(15 suppl):511. doi:10.1200/JCO.2017.35.15_suppl.511
  • Swain SM, Ewer MS, Viale G, et al. Pertuzumab, trastuzumab, and standard anthracycline- and taxane-based chemotherapy for the neoadjuvant treatment of patients with HER2-positive localized breast cancer (BERENICE): a Phase II, open-label, multicenter, multinational cardiac safety study. Ann Oncol. 2018;29(3):646–653. doi:10.1093/annonc/mdx773
  • Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(1):25–32. doi:10.1016/S1470-2045(11)70336-9
  • Fernandez-Martinez A, Krop IE, Hillman DW, et al. Survival, pathologic response, and genomics in CALGB 40601 (Alliance), a neoadjuvant Phase III trial of paclitaxel-trastuzumab with or without lapatinib in HER2-positive breast cancer. J Clin Oncol. 2020;38(35):4184–4193. doi:10.1200/JCO.20.01276
  • Swain SM, Baselga J, Kim SB, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015;372(8):724–734. doi:10.1056/NEJMoa1413513
  • Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783–1791. doi:10.1056/NEJMoa1209124
  • Saura C, Oliveira M, Feng YH, et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with ≥ 2 HER2-directed regimens: Phase III NALA trial. J Clin Oncol. 2020;38(27):3138–3149. doi:10.1200/JCO.20.00147
  • Smyth LM, Piha-Paul SA, Won HH, et al. Efficacy and determinants of response to HER kinase inhibition in HER2-mutant metastatic breast cancer. Cancer Discov. 2020;10(2):198–213. doi:10.1158/2159-8290.CD-19-0966
  • Warwas KM, Meyer M, Gonçalves M, et al. Co-stimulatory bispecific antibodies induce enhanced T cell activation and tumor cell killing in breast cancer models. Front Immunol. 2021;12:719116. doi:10.3389/fimmu.2021.719116
  • Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348(6230):74–80. doi:10.1126/science.aaa6204
  • Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;34(21):2460–2467. doi:10.1200/JCO.2015.64.8931
  • Adams S, Schmid P, Rugo HS, et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann Oncol. 2019;30(3):397–404. doi:10.1093/annonc/mdy517
  • Dirix LY, Takacs I, Jerusalem G, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN Solid Tumor study. Breast Cancer Res Treat. 2018;167(3):671–686. doi:10.1007/s10549-017-4537-5
  • Tolaney SM, Kalinsky K, Kaklamani V, et al. Phase 1b/2 study to evaluate eribulin mesylate in combination with pembrolizumab in patients with metastatic triple-negative breast cancer [abstract]. Cancer Res. 2018;78(Suppl):PD6–PD13. doi:10.1158/1538-7445.SABCS17-PD6-13
  • Adams S, Diamond JR, Hamilton E, et al. Atezolizumab plus nab-paclitaxel in the treatment of metastatic triple-negative breast cancer with 2-year survival follow-up: a phase 1b clinical trial. JAMA Oncol. 2019;5:334–342. doi:10.1001/jamaoncol.2018.5152
  • Schmid P, Rugo HS, Adams S, et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020;21(1):44–59. doi:10.1016/S1470-2045(19)30689-8
  • Miles D, Gligorov J, André F, et al. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol. 2021;32(8):994–1004. doi:10.1016/j.annonc.2021.05.801
  • Cortes J, Cescon DW, Rugo HS, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396(10265):1817–1828. doi:10.1016/S0140-6736(20)32531-9
  • Loi S, Giobbie-Hurder A, Gombos A, et al; International Breast Cancer Study Group and the Breast International Group. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b-2 trial. Lancet Oncol. 2019;20(3):371–382. doi:10.1016/S1470-2045(18)30812-X
  • Emens LA, Esteva FJ, Beresford M, et al. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised, double-blind trial. Lancet Oncol. 2020;21(10):1283–1295. doi:10.1016/S1470-2045(20)30465-4
  • Nanda R, Liu MC, Yau C, et al. Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing Phase 2 adaptively randomized I-SPY2 trial. JAMA Oncol. 2020;6(5):676–684. doi:10.1001/jamaoncol.2019.6650
  • Schmid P, Cortes J, Dent R, et al. KEYNOTE-522: phase 3 study of pembrolizumab (pembro) 1 chemotherapy (chemo) vs placebo (pbo) 1 chemo as neoadjuvant treatment, followed by pembro vs pbo as adjuvant treatment of early triple-negative breast cancer (TNBC). Ann Oncol. 2019;30(Suppl 5):v851–v934. doi:10.1093/annonc/mdz394.003
  • Loibl S, Untch M, Burchardi N, et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol. 2019;30(8):1279–1288. doi:10.1093/annonc/mdz158
  • Mittendorf EA, Zhang H, Barrios CH, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020;396(10257):1090–1100. doi:10.1016/S0140-6736(20)31953-X
  • Dastjerd NT, Valibeik A, Rahimi Monfared S, et al. Gene therapy: a promising approach for breast cancer treatment. Cell Biochem Funct. 2022;40(1):28–48.
  • Pandha HS, Martin LA, Rigg A, et al. Genetic prodrug activation therapy for breast cancer: a phase I clinical trial of erbB-2-directed suicide gene expression. J Clin Oncol. 1999;17(7):2180–2189.
  • Sun K, Xu Y, Zhang L, et al. A Phase 2 trial of enhancing immune checkpoint blockade by stereotactic radiation and in situ virus gene therapy in metastatic triple-negative breast cancer. Clin Cancer Res. 2022;28(20):4392–4401.