Skip to main content
Karger Author's Choice logoLink to Karger Author's Choice
. 2022 Aug 19;45(11):681–692. doi: 10.1159/000526481

Current Research Status of Metronomic Chemotherapy in Combination Treatment of Breast Cancer

Jiaxuan Liu 1, Maiyue He 1, Zijing Wang 1, Qiao Li 1,*, Binghe Xu 1,**
PMCID: PMC9677858  PMID: 35988534

Abstract

Background

Metronomic chemotherapy (MCT), termed sustained low-dose administration with minimal toxicity, is a new modality of conventional chemotherapy, a verified therapy alternative, and has acquired significant recognition and interest in oncology. Numerous clinical trials of MCT in combination with other treatments, including targeted therapies, biologics, and endocrine therapy, are in progress to obtain better results.

Summary

We comprehensively described the clinical benefits of MCT in combination with other treatments in different molecular subtypes of breast cancer and assessed the feasibility of its adoption in varying phases of treatment. Due to the promising preclinical and clinical investigations, it is expected that MCT in combination with other treatments will enhance the advantages of this strategy and apply it to clinical practice.

Key Message

MCT, in combination with other therapeutic interventions, will fully exploit the benefits of this strategy, ushering in a new paradigm in oncology treatment and driving the transformation of cancer into a more manageable chronic disease using newly developed treatment approaches.

Keywords: Breast cancer, Combination treatment, Metronomic chemotherapy, Molecular subtype

Introduction

From the early 20th century, when chemotherapy was recommended for treating cancer, to this day, when targeted therapies and immunotherapy medications are impacting cancer treatment, chemotherapy has remained the foundation of drug treatment for many cancers [1]. Conventional systematic chemotherapy hinges on the maximum tolerated dose (MTD) schedule, such that the maximum tolerable dose of chemotherapeutic agents is given in cycles with long intervals. However, because the traditional MTD schedule does not distinguish between malignant and healthy reproducing cells, it leads to unavoidable harmful impacts. Moreover, therapy-resistant tumor cells would regenerate between the drug-free periods, resulting in disease progression or regression. Therefore, alternative approaches to conventional chemotherapy have been investigated for years to enhance extended results and treatment adherence. The concept of metronomic chemotherapy (MCT) was initially introduced in 2000 [2, 3, 4], and it implies the sustained use of very low-dose chemotherapeutic agents with no prolonged drug-free period [4]. With this mode of administration, the drug is maintained at a reduced but efficient concentration for an extended period to exert antitumor action and reduce adverse effects while slowing the duration of drug resistance. The global emergence of a novel coronavirus (COVID-19) in December 2019 severely disrupted social orientation. As a result, the traditional diagnosis and treatment of tumors were dramatically hampered. Clinicians have compiled an urgent protocol to provide scientific and efficient alternatives for the management of breast cancer during the epidemic [5], among which metronomic chemotherapy is especially beneficial due to its unique advantages that vary from conventional chemotherapy, such as decreased infection risks because of lowered impact on patients' immunity, decreased hospital admittance, and improved quality of life.

MCT was first suggested as a complementary protocol when other tumor treatments proved ineffective. As a result, a plethora of investigations from the mid-2000s has been done to ascertain the usefulness of MCT in a range of cancers [6], particularly breast cancer [7, 8, 9], the most prevalent malignant tumor among women. With enhanced clinical benefits from earlier research, clinicians are starting to introduce MCT at varying phases of cancer treatment with intent to explore a deepened and more diverse application [10, 11, 12]. To obtain promising and improved clinical outcomes, a plethora of clinical trials of MCT in combination with other treatments, including targeted therapies, biologics, and endocrine therapy, are now in progress. In this review, we performed a systemic summary of published clinical studies to examine the clinical benefits of combining MCT with other treatments in different molecular subtypes of breast cancer. It also explored the feasibility of its adoption in varying phases of treatment. We researched the databases of PubMed/Medline, ClinicalTrials.gov, and the European Clinical Trials Database (EudraCT) for the key terms related to the topic. Only research papers published in English were considered, and registered ongoing clinical trials were also included. Case reports and reviews were excluded from our search. In addition, only the most recent version was included in the analysis for trials with multiple publications.

Mechanisms of Action − A Multi-Targeted Therapy

Initially thought to be an antiangiogenic treatment, metronomic chemotherapy has been advanced to enhance its anticancer effects through numerous inter-countered mechanisms, which distinguish it from conventional chemotherapy [10]. Angiogenesis, or the development of new blood vessels, is essential for the growth and metastasis of malignant tumors. In the traditional MTD schedule of cytotoxic agents, nonmalignant cells, particularly endothelial cells, may be recovered during the drug-free periods, leading to neo-angiogenesis, tumor growth, and eventually, metastasis. Clinical investigations have shown that traditional cytotoxic drugs have an antiangiogenic effect when administrated on a low-dose, consistent schedule, known as MCT [2, 13, 14]. It has been reported that activated endothelial cells are selectively sensitive to incredibly low and continuous dosages; therefore, MCT directly inhibits proliferation and/or induces apoptosis of activated endothelial cells [13]. The angiogenic transition in malignancy depends on the interference of pro- and antiangiogenic equilibrium, primarily in favor of pro-angiogenic factors and the recruitment of circulating endothelial progenitors (CEPs). Protracted exposure to reduced concentration agents also removes the additional angiogenesis stimulators derived from the tumor-supporting stroma. Furthermore, it improves the expression of a determined angiogenesis inhibitor, TSP-1, indirectly exerting an antiangiogenic effect [15, 16]. As a result of the decreased level and efficiency of bone marrow-derived CEPs generated by MCT, systematic angiogenesis is suppressed [15].

Severe immune suppression is common in malignant tumors, which facilitates immune evasion and subsequent tumor development. MCT potentiates host immunity. Regulatory T cells (Treg) are key suppressors of antitumor immune responses, and Treg accumulation in tumors is associated with a poor prognosis. Treg cells-selective depletion and peripheral T and natural killer (NK) effectors restoration were identified in patients with end-stage cancer exposed to metronomic cyclophosphamide (CTX) regimen to diminish the tumor-induced immunological tolerance [17]. It was discovered that low-dose chemotherapy exerts a direct immunostimulatory effect on dendritic cells (DCs) by modulating surface molecules [18], the enhanced maturation and functionality of DCs [19], and the increased antigen presentation to cytotoxic T lymphocytes via increased major histocompatibility complex (MHC) class I expression. Myeloid-derived suppressor cells (MDSCs) are targeted by low-dose chemotherapy and are reported to modulate the immune system [20, 21, 22].

Malignant tumors are reinforced by an identified stroma, which plays a crucial role in promoting tumor growth, progression, and aggression. The course comprises the infiltration of stromal cells, such as carcinoma-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), MDSCs, and the secretion of paracrine factors. Additionally, when standard chemotherapy is used, a chemo-modified process of the aforementioned stromal cells and secreted chemicals can negatively impact therapeutic responses, leading to the development of microenvironmental resistance [23, 24]. Conversely, a low-dose schedule of MCT substantially prevents therapy-induced alterations of tumor stroma, thus improving the treatment response and prolonging survival [25], demonstrating the viability of reversing drug resistance by changing the chemotherapy dosing schedule.

Clinical Trials and Practice in Various Breast Cancer Subtypes

HER2-Positive Breast Cancer

Roughly 20–25% of women with breast cancer have amplified or overexpressed Human Epidermal Growth Factor Receptor 2 (HER2) gene, which is associated with an increased risk of recurrence and poor prognosis [26]. Patients with HER2-positive breast cancer benefit from the introduction of HER2-targeted therapy, such as trastuzumab (TZM) and pertuzumab. Nevertheless, more than half of those individuals do not respond to those anti-HER2 monoclonal antibodies, and those who do will inevitably develop resistance within 1 year [27, 28]. Preclinical investigations suggested that anti-HER2 monoclonal antibodies could impede angiogenesis and tumor proliferation by regulating pro- and antiangiogenic factors, suggesting that they could be a simple alternative to combination antiangiogenic therapies, such as MCT [29, 30]. Furthermore, enhanced angiogenic actions via vascular endothelial growth factor (VEGF) upregulation in tumor resistance to TZM were reported in the murine model [31]. The authors also suggested that MCT could impede or reverse TZM resistance in HER2-positive breast cancer patients. Table 1 presents a summary of ongoing and accomplished preclinical studies and clinical trials in HER2-positive breast cancer. A preclinical study performed by Francia et al. [32] initially validated the survival prolongation and lesser toxicity of metastatic cancer in the context of TZM in combination with metronomic CTX.

Table 1.

Current and ongoing trials focusing metronomic chemotherapy in combination with other treatments in HER2-positive breast cancer

Setting Study, year Treatment regimen Phase Patients characteristics Study design Outcome Notes
Preclinical Francia et al. (2009) [32] mCTX + TZM Two HER-2 positive breast cancer models with established visceral metastases TZM 20 mg/kg i.p. twice/w + CTX 20 mg/kg p.o. qd versus MTD CTX 70 mg/kg i.p. D1, D3, D5 q21d (cumulative dose, 210 mg/kg/21 days) Survival prolongation of metastatic disease in the setting of mCTX + TZM, with lesser toxicity. As maintenance regimen following MTD-based protocol. Empirical 30% reduction MTD CTX dosing

Neoadjuvant Petry et al. (2015) [33] (TraQme trial) (NCT01329640) Doxorubicin + mCTX + TZM II 9 patients enrolled: untreated, locally advanced stage III HER-2-positive breast cancer (≥T2, ≥N1), no distant metastasis, no previous chemo or endocrine therapy. P 100 mg/m2 q1w*8w + TZM 2 mg/kg q1w (4 mg/kg initial dose) −> Doxorubicin 24 mg/m2 q1w + CTX 100 mg p.o. qd*9w + TZM 2 mg/kg q1w Survival: pCR rate: 55% (5/9); median follow-up: 33.6 months (5 alive/3 systemic disease recurrence/1 died due to disease progression). Toxicity: General mild hematological toxicity, no grade 4 event, unexpected grade 3 pneumonitis (2/9).

Arai et al. (2018) [34] Plasma level of MCSF and VEGF Stable MCSF level and downregulation of proangiogenic factor VEGF

Orlando et al. (2006) [35] TZM + m (MTX + CTX) II 22 patients: histologically confirmed HER2+ metastatic breast carcinoma TZM loading 8 mg/kg > 6 mg/kg i.v. q3w + (MTX 2.5 mg Bid d1, d4 + CTX 50 mg qd d1–d7) p.o. qw Survival: confirmed significant activity; 18% overall response (4/22), 46% clinical benefit (CR + PR + SD after 24 weeks). Toxicity: mild and well-tolerated; most frequent AE; 14% grade II leukopenia and 36% (8/22) increase in transaminase value (2 with grade III).

Orlando et al. (2020) [36] TZM + m (CAPE + CTX) II 60 patients: HER2+ breast cancer at first relapse or with synchronous metastasis TZM 4 mg/kg i.v. biweekly + (CAPE 1500 mg + CTX 50 mg) p.o. qd Survival: 56.7% (95% CI, 44.1–68.4%) ORR, 78.2% CBR; Median PFS 11 months, 1-year PFS 47.7%, median OS 45.9 months. Toxicity: excellent tolerability.

Wildiers et al. (2018) [37, 38] (EORTC 75111-10114) (NCT01597414) Dual anti-HER2 + mCTX II 80 patients (39/41): histologically proven HER2+ metastatic breast cancer, no previous chemotherapy for metastatic disease, ≥70 years or ≥60 years with functional restriction TZM loading 8 mg/kg −> 6 mg/kg i.v. q3w + pertuzumab loading 840 mg −> 420 mg i.v. q3w±CTX 50 mg p.o. qd Survival: 6-month PFS 46.2% (95% CI, 30.2–60.7) versus 73.4% (95% CI, 56.6–84.6), median PFS 5.6 months (95% CI, 3.6–16.8) versus 12.7 months (95% CI, 6.7–24.8), 1-year OS 67.3% (95% CI, 49.4–80.0) versus 83.8% (67.3–92.4), tumor response 44% (16/36) versus 53% (19/36); median follow-up: 54.0 months; Toxicity: limited additional toxicities.

Wang et al. (2021) [39] TZM + mNVB II 20 patients: HER2+ metastatic breast cancer with a median of 1 prior chemotherapy regimens TZM loading 8 mg/kg −> 6 mg/kg i.v. q3w + NVB 40 mg p.o. tiw Survival: 20% ORR, 75% CBR (4 PR + 11 SD), median PFS 7.4 months (95% CI, 3.2–11.5), median OS 45.8 months (95% CI, not reached); Toxicity: No grade 3/4 adverse events.

Saura et al. (2014) [40] (NCT00741260) Neratinib
(irreversible pan-TKI against HER1, HER2 and HER4) +CAPE
II 72 patients enrolled: confirmed HER-2 amplified metastatic or locally advanced breast cancer, progressed during or after more than 1 prior TZM-containing regimen and received prior taxane treatment, no prior capecitabine, anthracyclines or any other HER2-targeted therapy. (65 lapatinib naïve and 7 prior lapatinib pretreated patients) Neratinib 240 mg qd p.o. + CAPE 750 mg/m2 bid p.o. d1–d14 q21d Antitumor activity: 63% clinical responses in total: 64% (39/61) ORR, 72% CBR and median PFS 40.3 weeks in lapatinib-naïve patients and 57% (4/7) ORR, 71% CBR and median PFS 35.9 weeks in lapatinib pretreated patients. Safety: 10% treatment-related severe AEs; 26% grade 3/4 diarrhea, 14% PPE, 4% asthenia and 4% vomiting.

Ongoing NCT01873833 TZM + Metronomic (CAPE + CTX + Lapatinib) II Estimated 10 patients enrolled: histologically confirmed HER2+ metastatic breast cancer with prior TZM use in metastatic or adjuvant setting. No prior CAPE or lapatinib treatment and no more than two prior cytotoxic chemotherapeutic regimens for metastatic breast cancer. Capacitabine qd p.o. + CTX qd p.o. + Lapatinib qd p.o. + TZM d1 i.v.q21d

NCT03923166 Pyrotinib + Metronomic CAPE II Estimated 35 patients enrolled: pathologically confirmed HER2-expressing patients with locally advanced or metastatic breast cancer; progression after treatment with TZM Pyrotinib 400 mg qd p.o. + CAPE 500 mg tid p.o.

HER2, human epidermal growth factor receptor 2; CTX, cyclophosphamide; TZM, trastuzumab; MTD, maximum tolerated dose; pCR, pathologic complete response; ORR, objective response rate; CBR, clinical benefit rate; PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; SD, stable disease; CAPE, capecitabine; PPE, palmar-plantar erythrodysesthesia syndrome.

Multiple phase II trials evaluated the administration of HER2-targeted drugs in combination with MCT in metastatic settings, which provided a dependable alternative for later-line treatment. The combination [35] of TZM with a dual metronomic regimen, methotrexate (MTX) and CTX, has shown great efficacy in metastatic HER2-positive breast cancer and provided clinical benefit in a considerable fraction of patients who acquired TZM-resistance. To explore the feasibility of this strategy in first-line treatment of advanced HER2-positive breast cancer, a phase II trial of the Gruppo Oncologico Italia Meridionale (GOIM) was carried out by Orlando et al. [36]. Patients at first relapse or with synchronous metastasis were treated with TZM with metronomic capecitabine (CAPE) and CTX, which showed good clinical activity and excellent tolerability, with an objective response rate (ORR) of 56.7% (95% CI, 44.1–68.4%) and a clinical benefit rate (CBR) of 78.2% were presented. In the EORTC 75111-10114 trial [37], 80 patients with metastatic HER2-positive breast cancer were assigned to first-line treatment with dual anti-HER2 agents (TZM + pertuzumab) and metronomic oral CTX (50 mg Qd), with a 27.2% improvement in median progression-free survival compared to dual anti-HER2 treatment alone and an acceptable safety profile. Furthermore, second-line application of trastuzumab emtansine following disease progression in this context [37] was demonstrated to be efficient and well-tolerated, which appears to impede the need or exceed the application of conventional taxane-based chemotherapy. The long-term outcome data [38] from this trial recently presented a median follow-up of 54.0 months and demonstrated metronomic chemotherapy-based dual blockade as an active and relatively well-tolerated treatment option in the older or frail population. A single-arm phase II clinical trial carried out by Wang et al. [39] firstly verified the antitumor potency and well-tolerated toxicity of TZM and metronomic vinorelbine combination treatment in HER2-positive metastatic breast cancer patients. An ORR of 20.0% and a CBR of 75.0% were identified. Besides anti-HER2 monoclonal antibodies, the likelihood of combining MCT with tyrosine kinase inhibitors (TKIs) has been investigated. Saura et al. [40] reported a median progression-free survival (PFS) of 40.3 weeks in lapatinib-naïve HER2-positive breast cancer patients and 35.9 weeks in lapatinib-pretreated patients in the schedule of combining metronomic CAPE with neratinib, a potent irreversible pan-tyrosine kinase inhibitor.

The effectiveness and safety of MCT in breast cancer neoadjuvant treatment are also being verified progressively. A phase II clinical trial named TraQme [33] was allocated to patients with untreated, locally advanced stage III HER2-positive breast cancer, combining doxorubicin, TZM, and metronomic CTX, followed by paclitaxel and TZM in the preoperative therapy duration, with a 55.5% pathologic complete response (pCR) rate and a median follow-up of 33.6 months, was observed. Additional studies were conducted to ascertain the association of angiogenesis machinery between metronomic and TZM in a neoadjuvant context, and the downregulation of the proangiogenic factor VEGF [34] was detected in the combination treatment.

Patients with HER2-positive breast cancer, where prolonged treatment with anti-HER2 targeted drugs is feasible and safe, could gain from a combination with metronomic drugs to permit the extended distribution of the combination in different phases of treatment. Ongoing clinical trials are investigating different combinations to realize the maximum clinical benefit. A combination (NCT01873833) of TZM with metronomic agents (CAPE + CTX + Lapatinib) was investigated in HER2-positive metastatic breast cancer with prior TZM applied in the metastatic or adjuvant setting with anticipation. Pyrotinib, a newly-developed TKI in China, has been proven effective when administered with CAPE and has become the second-line standard treatment for HER2-positive metastatic breast cancer [41]. A single-arm, interventional, phase II clinical trial (NCT03923166) conducted by Li et al. [42] aims to investigate the efficacy and safety of pyrotinib in combination with metronomic CAPE in HER2-positive advanced breast cancer.

HR-Positive HER2-Negative Breast Cancer

Combining hormonal therapy and chemotherapy in patients with hormone receptor (HR) positive breast cancer began in the 1970s to improve therapeutic efficacy. Nevertheless, conventional chemotherapy is not administered in conjugation with endocrine therapy. The effectiveness of traditional MTD-scheduled chemotherapy primarily relies on the proliferative activity of malignant cells, while endocrine therapy is cytostatic [43]. A large randomized clinical trial validated the hypothesis of an antagonistic interface between two treatments [43, 44], and increased rates of adverse events are also reported in concurrent chemo-endocrine therapy treatment [45]. Conversely, MCT attacks the vascular compartments rather than tumor cells directly, and it is thought that combining it with this treatment could potentiate the effectiveness of endocrine therapy [46]. Furthermore, because a higher VEGF level has been linked to reduced responsiveness to antihormonal regimens [47], hypothetically, antiangiogenic metronomic treatment could prevent or delay hormonal treatment resistance. In breast cancer cell lines and animal models, combined endocrine treatment with a metronomic-like therapy showed improved antitumor action compared with either monotherapy [48, 49]. Table 2 presents a summary of ongoing and accomplished preclinical studies and clinical trials in HR-positive and HER2-negative breast cancer.

Table 2.

Current and ongoing trials focusing metronomic chemotherapy in combination with other treatments in HR-positive HER2-negative breast cancer

Setting Study, year Treatment regimen Phase Patients characteristics Study design Outcome Notes
Neoadjuvant Bottini et al. (2006) [46] LTZ (AI) versus LTZ + mCTX II 114 (104) patients: Elderly women (age >70) and women between 65 and 70 with clinical T2–4 N0–1 and ER+ and/or PR+ breast cancer unfit for chemotherapy LTZ 2.5 mg qd p.o. versus LTZ 2.5 mg qd p.o. + mCTX 50 mg qd p.o. for 6 months− > definitive surgery Combination of LTZ + mCTX is associated with a 2.79 increased odds (95% CI, 1.05–7.42) of response when compared with LTZ alone (p = 0.04), ORR: 71.9% (95% CI, 60.8–83.8) in LTZ arm versus 87.7% (95% CI, 78.6–96.2) in LTZ + mCTX arm

Ueno et al. (2018) [50, 51] (JBCRG-07) (UMIN000001331) LTZ (AI) + mCTX II 41 patients: previously untreated, clinical T2–4 N0–1 and ER-positive breast carcinoma, patients with postmenopausal status and ≥60 years LTZ 2.5 mg qd p.o. + mCTX 50 mg qd p.o. for 24 weeks −> surgical therapy after 1–4 weeks Outcomes: CRR: 67.5%, is associated with improved DFS (p = 0.020). Breast conserving rate: 75% (30/40).
Safety: Grade 3 or greater nonhematological toxicity was not reported. CECs: Patients with higher CEC counts at baseline or post-treatment showed worse DFS than those with lower counts (p < 0.001 at baseline and = 0.014 post-treatment).
Autophagy-related markers: Increase of autophagy-related markers (beclin 1 and LC3) and apoptosis-related markers (TUNEL and M30) following metronomic chemo-endocrine therapy and were associated with clinical response.

Generali et al. (2015) [52] Primary LTZ + mCTX versus LTZ alone II 107 women with T2–4 N0–1 and ER-positive breast cancer LTZ 2.5 mg qd p.o. versus LTZ 2.5 mg qd p.o. + mCTX 50 mg qd p.o. for 6 months HIF-1α reduction occurred in both the LTZ arm (45%) (n = 36/80) (p = 0.05) and the LTZ-CTX arm (55%) (n = 44/80) (p = 0.04) and was positively correlated with phospho-mTOR reduction after treatment (p < 0.03).

Bazzola et al. (2015) [53] LTZ + mCTX versus LTZ + mCTX + sorafenib II 13 ER-positive, postmenopausal, T2–4, N0-1 breast cancer patients LTZ 2.5 mg qd p.o. + mCTX 50 mg qd p.o. + sorafenib 400 mg bid q5d p.o for 6 months −> definitive surgery Outcome: A clinical CR in 6/13 patients and a significant reduction in tumor size in all 13 patients (p = 0.005). A significant reduction in SUV uptake in all patients between baseline and 30 days of treatment (p = 0.015) and between baseline and definitive surgery (p = 0.0002). Safety: The most common drug-related grade 3/4 adverse events were skin rash (69.3%), hand-foot skin reaction (69.3%) and diarrhea (46.1%). Molecular expression: A significant reduction of Ki-67 (14 days compared with baseline, p < 0.00001; definitive surgery compared with baseline, p < 0.03), CD-31 (p = 0.01) and VEGF-A (p = 0.007) expression in response to treatment.

Adamo et al. (2019) [54] (SOLTI-1501 VENTANA) (NCT02802748) LTZ + oral mVNB 0 61 (57) patients enrolled: postmenopausal, previously untreated, histologically confirmed stage I-IIIA invasive HR+ HER2− breast cancer (primary tumor diameter >1 cm and clinical nodal status 0–1), no multicentric tumors or prior anticancer therapy 3 arms: (1) LTZ 2.5 mg qd p.o.; (2) mVNB 50 mg tiw1 p.o.; (3) LTZ 2.5 mg Qd + mVNB 50 mg 3d/w p.o. q21d. −> surgery after 3w Antiproliferative effect: mVNB + LTZ combination regimen (73.2%) was superior to both monotherapy arms combined (49.9%) or monotherapy alone (LTZ: 65.7%; mVNB: 19.1%). Safety: grade 3 adverse events occurred in 3.4% cases. Gene profile: combination regimen induced high expression of immune-related genes (CD8 T cell signature and PDL1 gene) and a statistically significant increase of sTILs in tumor with ≤10% sTILs at baseline

Metastatic Li et al. (2019) [42] mCAPE + AI (exemestane/LTZ) II 44 postmenopausal HR-positive advanced or metastatic breast cancer patients who exhibited disease progression after first-line AIs treatment and who could not tolerate or rejected conventional chemotherapy mCAPE 500 mg tid p.o. + exemestane 25 mg qd p.o./LTZ 2.5 mg qd p.o. Outcome: After a median follow-up of 14.8 months, ORR was 70.5%, CBR − 77.3%, PFS − 16.2 months, and TTF − 14.4 months. Safety: Grade 3 toxicities (hand–foot syndrome) were observed only in 4 of the patients. Most patients exhibited no or mild toxicities.

Aurilio et al. (2012) [55] Fulvestrant + mCTX + mMTX II 33 postmenopausal patients with heavily pretreated ER-positive advanced breast cancer. mCTX 50 mg qd p.o. + mMTX 25 mg bid biw1 p.o. + fulvestrant 250 mg q28d i.m. Median PFS: 4.7 months, prognosis was worse in patients who had already received many lines of therapy; median OS: 43.6 months.

Schwartzberg et al. (2014) [56] Fulvestrant + mCAPE II 41 women with histologically or cytologically confirmed, HER2–, ER+ and/or PR-positive metastatic breast cancer Fulvestrant: 500 mg d1, 250 mg d15&d29 p.o. −> 250 mg q28d + mCAPE: bid p.o. (dose depends on weight) Outcome: median PFS: 14.98 months, median TTP: 26.94 months, median OS: 28.65 months. CBR (CR + PR + SD ≥24 weeks): 58.5%. Safety: 13 (31.7%) patients who experienced any grade 3 events, but only 4 (9.8%) patients who experienced any grade 4 event. Only 2 patients discontinued therapy because of any treatment-related toxicity.

Ongoing NCT04571437 (B-001 Study) LTZ + mCAPE versus LTZ alone II Estimated 204 patients enrolled: ER-positive and HER2-negative breast cancer proved by IHC, with metastatic/recurrent diseases LTZ 2.5 mg qd p.o. + mCAPE versus LTZ 2.5 mg qd p.o.

NCT05063136 mCAPE + endocrine therapy III Estimated 1,979 patients enrolled: Invasive breast cancer patients with HR (+) and HER2(−) CAPE 500 mg tid p.o. + standard endocrine therapy Adjuvant therapy

HR, hormone receptor; HER2, human epidermal growth factor receptor 2; LTZ, letrozole; AI, aromatase inhibitor; CTX, cyclophosphamide; ER, estrogen receptor; PR, progesterone receptor; ORR, objective response rate; CRR, clinical response rate; DFS, disease-free survival; CEC, circulating endothelial cell; CR, complete response; SUV, standard uptake value; VNB, vinorelbine; sTILs, stromal tumor infiltrating lymphocytes; CAPE, capecitabine; CBR, clinical benefit rate; PFS, progression-free survival; TTF, time to treatment failure; OS, overall survival; MTX, methotrexate; TTP, time to progression; CR, complete response; PR, partial response; SD, stable disease.

Endocrine therapy is a crucial constituent of the systematic treatment of postmenopausal patients with HR-positive breast cancer, which is preferred to neoadjuvant treatment because of its lower toxicity than chemotherapy. Cancer treatment with aromatase inhibitors, such as letrozole, improves the response rate to conventional tamoxifen in breast cancer patients [57]. To further improve surgical outcomes, it is crucial to enhance the response rate of neoadjuvant endocrine therapy without significant adverse events. In this context, combining MCT with a neoadjuvant endocrine treatment may have specific benefits, with oral CTX being one of the most commonly used metronomic agents. The possibility of combined administration of aromatase inhibitor letrozole with metronomic-scheduled CTX in the neoadjuvant therapy of elderly patients with breast cancer has been investigated earlier [46]; an ORR of 87.7% was reported in patients treated with letrozole plus metronomic CTX, whereas an ORR of 71.9% was reported in patients treated with letrozole alone. Additionally, a considerably reduced posttreatment expression of Ki-67 was observed in the combination treatment compared with letrozole monotherapy. A multicenter phase II clinical trial (JBCRG-07: UMIN000001331) [50, 51] of neoadjuvant metronomic chemo-endocrine therapy with letrozole and CTX was following conducted in postmenopausal, T2−4 N0−1, and estrogen receptor-positive breast carcinoma. The clinical response rate was 67.5%, which demonstrated a link with improved disease-free survival (DFS) (p = 0.020). As for the surgical outcome, the breast-conserving rate was 75%, and 18 of 28 (64%) who would have undergone total mastectomy before neoadjuvant chemo-endocrine treatment subsequently underwent breast-conserving surgery. Enhanced safety was reported, with no grade 3 or advanced non-hematological adverse events, and no treatment discontinuation because of adverse events. The subject group additionally confirmed the independent prognostic efficacy of circulating endothelial cell (CEC) counts [50] and the modulating effect of metronomic chemo-endocrine treatment on factors related to autophagy and apoptosis [51]. Generali et al. [52] also found a letrozole-based metronomic treatment regulated mTOR and HIF-1α expression, which is associated with clinical response in the neoadjuvant population. Previous studies have described the underlying mechanisms of the resistance to endocrine treatment and the prospects of combining endocrine treatment with signal-transduction inhibitors to evade resistance and improve clinical response [58, 59]. Sorafenib is an oral multiple kinase inhibitor capable of inactivating numerous kinases and signaling pathways involved in tumor development and aggression. Bazzola et al. [53] planned a phase II clinical trial to study the action of the administrative combination of letrozole, metronomic CTX, and sorafenib with enhanced antiangiogenic efficiency. A clinical complete response (CR) was observed in 6/13 patients, and a significant reduction in standard uptake value (SUV) uptake in all patients following treatment was verified. The tolerable toxicity and regulation of specific biomarker expression additionally indicated the activity of the combined treatment. The SOLTI-1501 VENTANA trial [54] assessed the antiproliferative impact and tolerance of oral metronomic vinorelbine in combination with endocrine treatment in patients with untreated HR+/HER2− breast cancer. A higher expression of immune-related genes and stromal tumor-infiltrating lymphocytes (sTILs) observed in the combined treatments presented a basis for this combination with immunotherapy.

HR-positive metastatic breast cancer is particularly difficult to treat as patients treated with prior endocrine therapy including antiestrogen and aromatase inhibitors, develop resistance to traditional estrogen receptor (ER) blockade treatment. The underlying mechanisms of endocrine resistance have not yet been clarified. As a result, VEGF-regulated tumor angiogenesis may play a crucial function in the failure of first-line and second-line endocrine therapy in HR-positive breast cancer. A phase II clinical trial conducted by Li et al. [42] combined metronomic CAPE with aromatase inhibitors (exemestane or letrozole) in postmenopausal HR-positive advanced breast cancer patients that progressed after prior endocrine treatment or were intolerant to traditional chemotherapy. After a median follow-up of 14.8 months, this trial yielded an ORR of 70.5%, a CBR of 77.3%, a PFS of 16.2 months, and a time to treatment failure (TTF) of 14.4 months, while most patients exhibited mild or no toxicities. Fulvestrant is a recently developed selective ER antagonist that is efficient in preventing metastatic breast cancer progression after initial antiestrogen treatment [60]. A combination of metronomic CTX and MTX with fulvestrant in ER-positive advanced breast cancer was conducted by Aurilio et al. [55]. Antitumor activity and minimal toxicity of the combined treatment providing extended disease modulation reinforced this regimen as an efficient therapeutic tool. In another research, the combined administration of fulvestrant and metronomic CAPE in postmenopausal HR-positive, HER2-negative breast cancer patients yielded a median PFS of 14.98 months and a median time to progression (TTP) of 26.94 months, respectively [56]. The meaningful TTP with good tolerance presented reliable proof for metronomic chemo-endocrine therapy.

There are also several ongoing clinical trials investigating the combination of metronomic chemotherapy, such as CAPE or CTX, with endocrine therapy for the adjuvant or salvage treatment of HR-positive, HER2-negative breast cancer patients. The promising results would provide a strong basis for further use in clinical practice.

Triple-Negative Breast Cancer

Triple-negative breast cancer (TNBC) is a subtype of breast cancer that accounts for approximately 15% of all incidences and is characterized by high aggressiveness, early recurrence, and poor prognosis with limited therapeutic alternatives. Instead of expression of ER, progesterone receptor (PR), and HER2, TNBC has a unique molecular profile, including overexpression of VEGF and epidermal growth factor receptor (EGFR), which are involved in the angiogenesis modulation. As a result, there is a growing body of evidence supporting the use of antiangiogenic therapies in TNBC patients. The proposition that their antiangiogenic efficiency may be improved in combination with a metronomic schedule had been clarified earlier [61]. Table 3 summarizes the ongoing and accomplished preclinical studies and clinical trials in TNBC. In preclinical examinations, the potential therapeutic impact of continuous low-dose metronomic topotecan in simultaneous combination with an angiogenic TKI inhibitor presents a significant basis for treating metastatic TNBC patients [62]. Bevacizumab is one of the extensively used antiangiogenic agents, which is a recombinant humanized monoclonal antibody against VEGF. The combination of bevacizumab with the newly developed small-molecule tyrosine kinase inhibitor, erlotinib, demonstrated activities in pretreated metastatic breast cancer [67]. A phase II clinical trial was carried out by Montagna et al. [63] combining metronomic chemotherapy (CTX + CAPE) with antiangiogenic bevacizumab plus erlotinib in patients with HER2-negative and low HRs expression metastatic breast cancer. The combined treatment was well tolerated and efficient with a median clinical benefit of 75% and median TTP of 43 weeks. The special molecular properties of TNBC also involve a high rate of deleterious BRCA gene mutation, which participates in the DNA impairment procedures and is sensitive to poly [ADP-ribose] polymerase (PARP) inhibition. With the properties of induction of DNA impairment, we recorded the clinical advantages of metronomic chemotherapy in combination with a PARP inhibitor [68]. As patients carrying BRCA mutations are highly prone to develop TNBC [69], researchers set out to examine the activity of this combination in patients with TNBC [64, 65]. Unfortunately, the combination of metronomic CTX with PARP inhibitor Veliparib, at the dosage and schedule assessed, produced controversial results in the two trials; as a result, this warrants additional investigation. Researchers are going to evaluate the combination of tucidinostat, HDAC inhibitor, with metronomic CAPE that has been approved for metastatic triple-negative breast cancer that has failed at first-line taxane therapy in a randomized controlled phase clinical trial (NCT05390476).

Table 3.

Current and ongoing trials focusing metronomic chemotherapy in combination with other treatments in Triple negative breast cancer

Setting Study, year Treatment regimen Phase Patients characteristics Study design Outcome Notes
Combined with antiangiogenesis therapy Di Desidero et al. (2015) [62] Metronomic topotecan (topoI inhibitor) + pazopanib (TKI) Preclinical A triple-negative, primary and metastatic breast cancer orthotopic model Metronomic topotecan + pazopanib (TKI) A significantly enhanced antitumor activity and prolonged survival. A marked decrease in tumor vascularity, proliferation index, and the induction of apoptosis on both endothelial cells and TNBC cells.

Montagna et al. (2012) [63] Metronomic chemotherapy (CTX + CAPE) + erlotinib & bevacizumab (BEXE) II 24 patients with HER2-negative MBC and poor HR expression CTX 50 mg qd p.o. + CAPE 500 mg tid p.o. + erlotinib 100 mg qd p.o. + bevacizumab 15 mg/kg q21d i.v. Outcome: ORR: 62% (95% CI, 41–81%);
Overall CBR: 75% (95% CI, 53–90%); Median
TTP: 43 weeks (95% CI, 21–69 weeks), OS:
108 months (95% CI, 70–110).
Toxicity: Generally mild.
CEPs: low level of CEPs at baseline had a
significantly improved PFS

Combined with PARP inhibitors Anampa et al. (2018) [64] (NCT02824575) Metronomic CTX + PARP inhibitors (veliparib) I HER2-negative MBC CTX 50/75/100/125 mg qd p.o. + Veliparib 200 mg bid p.o. The combination is well tolerated and shows antitumor activity in patients with ΒRCA mutation associated MBC

Kummar et al. (2016) [65] (NCT01306032) Metronomic CTX + PARP inhibitors (veliparib) II 45 (39) metastatic TNBC patients disease had progressed following at least one line of standard therapy CTX 50 mg qd p.o. versus CTX 50 mg qd p.o. + Veliparib 60 mg qd p.o. Response rates and median PFS did not significantly differ between both treatment arms

Combined with HDAC inhibitor NCT05390476 (ongoing) Metronomic CAPE + tucidinostat (HDAC inhibitor) versus metronomic CAPE alone II Estimated 125 patients enrolled: histologically confirmed triple-negative metastatic breast cancer, metastatic breast cancer that has failed at first-line taxane therapy CAPE 500 mg tid p.o. + tucidinostat 20 mg d1/4/8/11/15/18 p.o. q3w versus CAPE 500 mg tid p.o.

Combined with immunotherapy Chen et al. (2020) [66] Monoclonal antibody (PD-1) + metronomic paclitaxel Preclinical TNBC mouse models PD-1 mAb alone versus paclitaxel alone versus PD-1 mAb 200 µg/kg q3d i.p. + paclitaxel 6 mg/kg q2d p.o. The combination regimen has a potent antitumor effect. Metronomic paclitaxel changed the immune cell population

NCT04389073 (ongoing) Toripalimab (anti-PD-1 antibody) + metronomic vinorelbine II HER2-negative metastatic breast cancer patients Toripalimab 240 mg d1 i.v. q3w + vinorelbine 40 mg/day p.o. tiw

TKI, tyrosine kinase inhibitor; TNBC, triple negative breast cancer; CTX, cyclophosphamide; CAPE, capecitabine; HER2, human epidermal growth factor receptor 2; MBC, metastatic breast cancer; HR, hormone receptor; ORR, objective response rate; CBR, clinical benefit rate; TTP, time to progression; OS, overall survival; CEPs, circulating endothelial progenitors; PFS, progression-free survival; PARP, poly [ADP-Ribose] polymerase; mAb, monoclonal antibody.

In recent years, immunotherapy has emerged as a promising discipline in oncology. Although, unlike other breast cancer subtypes, there is no specific targeted therapy for metastatic TNBC, the generally higher level of TILs and immune checkpoint molecules in TNBC makes it an attractive objective for immunotherapy [70, 71]. The PD-1/PD-L1 blockade yielded extended responses in metastatic TNBC; however, only a proportion of patients gained from the immunotherapy because of heterogeneous tumor immune microenvironments [72, 73, 74, 75]. In the TONIC trial [76], short-term or metronomic chemotherapy could induce a more favorable immune microenvironment and improve therapy sensitivity to PD-1 blockade in metastatic TNBC. Chue et al. [77, 78] described the use of consecutive metronomic chemotherapy combined with immunotherapy in 2 cases of recurrent metastatic TNBC patients, both of whom attained extended remission and produced a prospective alternative for the treatment. Furthermore, the combination treatment of metronomic paclitaxel with PD-1 monoclonal antibody (mAb) produced a powerful antitumor effect in preclinical TNBC mouse models [66]. This study also proposed that metronomic paclitaxel enhanced immunotherapy potency via immune microenvironment transformation. A single-arm, phase II clinical trial (NCT04389073) in HER2-negative metastatic breast cancer patients aims to evaluate the clinical activity of Toripalimab, anti-PD-1 antibody, in combination with metronomic vinorelbine. They also explore the combination strategy of metronomic chemotherapy with immunotherapy and antiangiogenesis, of which results are worth waiting for.

One of the recently developed immunotherapy alternatives is cancer vaccines, which induce special immunity to cancer antigens. Numerous studies have combined metronomic chemotherapy with vaccine immunotherapy in metastatic breast cancer. The potential outcome of combination treatment in improving the survival and quality of life of the patient has been shown [79, 80], providing ideas for combined treatment strategies.

Conclusion

MCT is known as sustained low-dose administration with minimal toxicity and is a new modality of administering conventional chemotherapy. It is a verified therapy option and has obtained significant recognition and interest in antitumor therapy. Due to the promising preclinical and clinical investigations, it is expected that MCT in combination with other treatments will enhance the advantages of this strategy and apply it to clinical practice.

This systematic review also reveals some critical thoughts. First, there is limited high quality data, such as phase III trials of the combination strategy of MCT with other treatments. Therefore, it would restrict the further approval and application in clinical guidelines and practice. We await the results of the relevant phase III clinical trials currently ongoing and expect more researchers to conduct more high-quality clinical trials. Moreover, the previous clinical trials mostly examined the combination strategy in breast cancer patients without defined molecular subtypes. With the further development of individualized treatment and precision medicine, combined therapeutic options targeted for specific populations will gain more benefits.

The combination strategy of MCT with other treatments is worth further investigation, which is hopefully forming a new paradigm in oncology treatment and promoting turning cancer into a more manageable chronic disease with newly developed therapeutic options.

Conflict of Interest Statement

The authors have no conflicts of interest to declare.

Funding Sources

The authors have no relevant financial or nonfinancial interests to disclose.

Author Contributions

J. Liu, Q. Li, and B. Xu contributed to the idea approval of this systemic review. Literature search and data analysis were performed by J. Liu, M. He, and Z. Wang. J. Liu drafted the manuscript and Q. Li critically revised the work. All the authors read and approved the final manuscript.

Funding Statement

The authors have no relevant financial or nonfinancial interests to disclose.

References

  • 1.DeVita VT, Jr, Chu E. A history of cancer chemotherapy. Cancer Res. 2008;68((21)):8643–8653. doi: 10.1158/0008-5472.CAN-07-6611. [DOI] [PubMed] [Google Scholar]
  • 2.Browder T, Butterfield CE, Kraling BM, Shi B, Marshall B, O'Reilly MS, et al. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res. 2000;60((7)):1878–1886. [PubMed] [Google Scholar]
  • 3.Klement G, Baruchel S, Rak J, Man S, Clark K, Hicklin DJ, et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. J Clin Invest. 2000;105((8)):R15–R24. doi: 10.1172/JCI8829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hanahan D, Bergers G, Bergsland E. Less is more, regularly metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J Clin Invest. 2000;105((8)):1045–1047. doi: 10.1172/JCI9872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Li L, Ma F. Interpretation of guidelines for the diagnosis and treatment of breast cancer during the COVID-19 outbreak. Chin J Clin Oncol. 2020;8((47)) [Google Scholar]
  • 6.Lien K, Georgsdottir S, Sivanathan L, Chan K, Emmenegger U. Low-dose metronomic chemotherapy a systematic literature analysis. Eur J Cancer. 2013;49((16)):3387–3395. doi: 10.1016/j.ejca.2013.06.038. [DOI] [PubMed] [Google Scholar]
  • 7.Cazzaniga ME, Torri V, Riva F, Porcu L, Cicchiello F, Capici S, et al. Efficacy and safety of vinorelbine-capecitabine oral metronomic combination in elderly metastatic breast cancer patients VICTOR-1 study. Tumori. 2017;103((1)):e4–e8. doi: 10.5301/tj.5000543. [DOI] [PubMed] [Google Scholar]
  • 8.Cazzaniga ME, Munzone E, Bocci G, Afonso N, Gomez P, Langkjer S, et al. Pan-European Expert Meeting on the use of metronomic chemotherapy in advanced breast cancer patients the PENELOPE project. Adv Ther. 2019;36((2)):381–406. doi: 10.1007/s12325-018-0844-4. [DOI] [PubMed] [Google Scholar]
  • 9.Krajnak S, Battista M, Brenner W, Almstedt K, Elger T, Heimes AS, et al. Explorative analysis of low-dose metronomic chemotherapy with cyclophosphamide and methotrexate in a cohort of metastatic breast cancer patients. Breast Care. 2018;13((4)):272–276. doi: 10.1159/000487629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.André N, Carré M, Pasquier E. Metronomics towards personalized chemotherapy? Nat Rev Clin Oncol. 2014;11((7)):413–431. doi: 10.1038/nrclinonc.2014.89. [DOI] [PubMed] [Google Scholar]
  • 11.Gatenby RA. A change of strategy in the war on cancer. Nature. 2009;459((7246)):508–509. doi: 10.1038/459508a. [DOI] [PubMed] [Google Scholar]
  • 12.Perroud HA, Alasino CM, Rico MJ, Queralt F, Pezzotto SM, Rozados VR, et al. Quality of life in patients with metastatic breast cancer treated with metronomic chemotherapy. Future Oncol. 2016;12((10)):1233–1242. doi: 10.2217/fon-2016-0075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4((6)):423–436. doi: 10.1038/nrc1369. [DOI] [PubMed] [Google Scholar]
  • 14.Gille J, Spieth K, Kaufmann R. Metronomic low-dose chemotherapy as antiangiogenic therapeutic strategy for cancer. J Dtsch Dermatol Ges. 2005;3((1)):26–32. doi: 10.1046/j.1439-0353.2005.04048.x. [DOI] [PubMed] [Google Scholar]
  • 15.Natale G, Bocci G. Does metronomic chemotherapy induce tumor angiogenic dormancy? A review of available preclinical and clinical data. Cancer Lett. 2018;432:28–37. doi: 10.1016/j.canlet.2018.06.002. [DOI] [PubMed] [Google Scholar]
  • 16.Bocci G, Francia G, Man S, Lawler J, Kerbel RS. Thrombospondin 1, a mediator. of the antiangiogenic effects of low-dose metronomic chemotherapy. Proc Natl Acad Sci U S A. 2003;100((22)):12917–22. doi: 10.1073/pnas.2135406100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother. 2007;56((5)):641–648. doi: 10.1007/s00262-006-0225-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Shurin GV, Tourkova IL, Kaneno R, Shurin MR. Chemotherapeutic agents in noncytotoxic concentrations increase antigen presentation by dendritic cells via an IL-12-dependent mechanism. J Immunol. 2009;183((1)):137–144. doi: 10.4049/jimmunol.0900734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tanaka H, Matsushima H, Nishibu A, Clausen BE, Takashima A. Dual therapeutic efficacy of vinblastine as a unique chemotherapeutic agent capable of inducing dendritic cell maturation. Cancer Res. 2009;69((17)):6987–6994. doi: 10.1158/0008-5472.CAN-09-1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Doloff JC, Chen CS, Waxman DJ. Anti-tumor innate immunity activated by intermittent metronomic cyclophosphamide treatment of 9L brain tumor xenografts is preserved by anti-angiogenic drugs that spare VEGF receptor 2. Mol Cancer. 2014;13:158. doi: 10.1186/1476-4598-13-158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kareva I. A combination of immune checkpoint inhibition with metronomic chemotherapy as a way of targeting therapy-resistant cancer cells. Int J Mol Sci. 2017;18((10)):2134. doi: 10.3390/ijms18102134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Peereboom DM, Alban TJ, Grabowski MM, Alvarado AG, Otvos B, Bayik D, et al. Metronomic capecitabine as an immune modulator in glioblastoma patients reduces myeloid-derived suppressor cells. JCI Insight. 2019;4((22)):e130748. doi: 10.1172/jci.insight.130748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med. 2012;18((9)):1359–1368. doi: 10.1038/nm.2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nakasone ES, Askautrud HA, Kees T, Park JH, Plaks V, Ewald AJ, et al. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell. 2012;21((4)):488–503. doi: 10.1016/j.ccr.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chan TS, Hsu CC, Pai VC, Liao WY, Huang SS, Tan KT, et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J Exp Med. 2016;213((13)):2967–2988. doi: 10.1084/jem.20151665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jackisch C, Lammers P, Jacobs I. Evolving landscape of human epidermal growth factor receptor 2-positive breast cancer treatment and the future of biosimilars. Breast. 2017;32:199–216. doi: 10.1016/j.breast.2017.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Piccart M. Circumventing de novo and acquired resistance to trastuzumab new hope for the care of ErbB2-positive breast cancer. Clin Breast Cancer. 2008;8((Suppl 3)):S100–S113. doi: 10.3816/cbc.2008.s.006. [DOI] [PubMed] [Google Scholar]
  • 28.Hudis CA. Trastuzumab mechanism of action and use in clinical practice. N Engl J Med. 2007;357((1)):39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  • 29.Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumour biology herceptin acts as an anti-angiogenic cocktail. Nature. 2002;416((6878)):279–280. doi: 10.1038/416279b. [DOI] [PubMed] [Google Scholar]
  • 30.Wen XF, Yang G, Mao W, Thornton A, Liu J, Bast RC, Jr, et al. HER2 signaling modulates the equilibrium between pro- and antiangiogenic factors via distinct pathways implications for HER2-targeted antibody therapy. Oncogene. 2006;25((52)):6986–6996. doi: 10.1038/sj.onc.1209685. [DOI] [PubMed] [Google Scholar]
  • 31.du Manoir JM, Francia G, Man S, Mossoba M, Medin JA, Viloria-Petit A, et al. Strategies for delaying or treating in vivo acquired resistance to trastuzumab in human breast cancer xenografts. Clin Cancer Res. 2006;12((3 Pt 1)):904–916. doi: 10.1158/1078-0432.CCR-05-1109. [DOI] [PubMed] [Google Scholar]
  • 32.Francia G, Man S, Lee CJ, Lee CR, Xu P, Mossoba ME, et al. Comparative impact of trastuzumab and cyclophosphamide on HER-2-positive human breast cancer xenografts. Clin Cancer Res. 2009;15((20)):6358–6366. doi: 10.1158/1078-0432.CCR-09-0931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Petry V, Gagliato DM, Leal AIC, Arai RJ, Longo E, Andrade F, et al. Metronomic chemotherapy in the neoadjuvant setting results of two parallel feasibility trials (TraQme and TAME) in patients with HER2+ and HER2- locally advanced breast cancer. Braz J Med Biol Res. 2015;48((5)):479–485. doi: 10.1590/1414-431X20144354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Arai RJ, Petry V, Hoff PM, Mano MS. Serum levels of VEGF and MCSF in HER2+/HER2- breast cancer patients with metronomic neoadjuvant chemotherapy. Biomark Res. 2018;6:20. doi: 10.1186/s40364-018-0135-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Orlando L, Cardillo A, Ghisini R, Rocca A, Balduzzi A, Torrisi R, et al. Trastuzumab in combination with metronomic cyclophosphamide and methotrexate in patients with HER-2 positive metastatic breast cancer. BMC Cancer. 2006;6:225. doi: 10.1186/1471-2407-6-225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Orlando L, Lorusso V, Giotta F, Di Maio M, Schiavone P, Fedele P, et al. Metronomic oral chemotherapy with cyclophosphamide plus capecitabine combined with trastuzumab (HEX) as first line therapy of HER-2 positive advanced breast cancer a phase II trial of the Gruppo Oncologico Italia Meridionale (GOIM) Breast. 2020;53:18–22. doi: 10.1016/j.breast.2020.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wildiers H, Tryfonidis K, Dal Lago L, Vuylsteke P, Curigliano G, Waters S, et al. Pertuzumab and trastuzumab with or without metronomic chemotherapy for older patients with HER2-positive metastatic breast cancer (EORTC 75111-10114) an open-label, randomised, phase 2 trial from the Elderly Task Force/Breast Cancer Group. Lancet Oncol. 2018;19((3)):323–336. doi: 10.1016/S1470-2045(18)30083-4. [DOI] [PubMed] [Google Scholar]
  • 38.Wildiers H, Meyskens T, Marreaud S, Lago LD, Vuylsteke P, Curigliano G, et al. Long term outcome data from the EORTC 75111-10114 ETF/BCG randomized phase II study Pertuzumab and trastuzumab with or without metronomic chemotherapy for older patients with HER2-positive metastatic breast cancer, followed by T-DM1 after progression. Breast. 2022;64:100–11. doi: 10.1016/j.breast.2022.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wang Z, Liu J, Ma F, Wang J, Luo Y, Fan Y, et al. Safety and efficacy study of oral metronomic vinorelbine combined with trastuzumab (mNH) in HER2-positive metastatic breast cancer a phase II trial. Breast Cancer Res Treat. 2021;188((2)):441–447. doi: 10.1007/s10549-021-06216-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Saura C, Garcia-Saenz JA, Xu B, Harb W, Moroose R, Pluard T, et al. Safety and efficacy of neratinib in combination with capecitabine in patients with metastatic human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2014;32((32)):3626–3633. doi: 10.1200/JCO.2014.56.3809. [DOI] [PubMed] [Google Scholar]
  • 41.Xu B, Yan M, Ma F, Hu X, Feng J, Ouyang Q, et al. Pyrotinib plus capecitabine versus lapatinib plus capecitabine for the treatment of HER2-positive metastatic breast cancer (PHOEBE) a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2021;22((3)):351–360. doi: 10.1016/S1470-2045(20)30702-6. [DOI] [PubMed] [Google Scholar]
  • 42.Li JW, Zuo WJ, Ivanova D, Jia XQ, Lei L, Liu GY. Metronomic capecitabine combined with aromatase inhibitors for new chemoendocrine treatment of advanced breast cancer a phase II clinical trial. Breast Cancer Res Treat. 2019;173((2)):407–415. doi: 10.1007/s10549-018-5024-3. [DOI] [PubMed] [Google Scholar]
  • 43.Osborne CK, Kitten L, Arteaga CL. Antagonism of chemotherapy-induced cytotoxicity for human breast cancer cells by antiestrogens. J Clin Oncol. 1989;7((6)):710–717. doi: 10.1200/JCO.1989.7.6.710. [DOI] [PubMed] [Google Scholar]
  • 44.Albain KS, Barlow WE, Ravdin PM, Farrar WB, Burton GV, Ketchel SJ, et al. Adjuvant chemotherapy and timing of tamoxifen in postmenopausal patients with endocrine-responsive node-positive breast cancer a phase 3, open-label, randomised controlled trial. Lancet. 2009;374((9707)):2055–2063. doi: 10.1016/S0140-6736(09)61523-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pritchard KI, Paterson AH, Paul NA, Zee B, Fine S, Pater J. Increased thromboembolic complications with concurrent tamoxifen and chemotherapy in a randomized trial of adjuvant therapy for women with breast cancer. National Cancer Institute of Canada Clinical Trials Group Breast Cancer Site Group. J Clin Oncol. 1996;14((10)):2731–2737. doi: 10.1200/JCO.1996.14.10.2731. [DOI] [PubMed] [Google Scholar]
  • 46.Bottini A, Generali D, Brizzi MP, Fox SB, Bersiga A, Bonardi S, et al. Randomized phase II trial of letrozole and letrozole plus low-dose metronomic oral cyclophosphamide as primary systemic treatment in elderly breast cancer patients. J Clin Oncol. 2006;24((22)):3623–3628. doi: 10.1200/JCO.2005.04.5773. [DOI] [PubMed] [Google Scholar]
  • 47.Manders P, Beex LVAM, Tjan-Heijnen VCG, Span PN, Sweep CGJ. Vascular endothelial growth factor is associated with the efficacy of endocrine therapy in patients with advanced breast carcinoma. Cancer. 2003;98((10)):2125–2132. doi: 10.1002/cncr.11764. [DOI] [PubMed] [Google Scholar]
  • 48.Nukatsuka M, Saito H, Nakagawa F, Abe M, Uchida J, Shibata J, et al. Oral fluoropyrimidine may augment the efficacy of aromatase inhibitor via the down-regulation of estrogen receptor in estrogen-responsive breast cancer xenografts. Breast Cancer Res Treat. 2011;128((2)):381–390. doi: 10.1007/s10549-010-1141-3. [DOI] [PubMed] [Google Scholar]
  • 49.Yang W, Hosford SR, Dillon LM, Shee K, Liu SC, Bean JR, et al. Strategically timing inhibition of phosphatidylinositol 3-Kinase to maximize therapeutic index in estrogen receptor alpha-positive PIK3CA-mutant breast cancer. Clin Cancer Res. 2016;22((9)):2250–2260. doi: 10.1158/1078-0432.CCR-15-2276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ueno T, Masuda N, Kamigaki S, Morimoto T, Akiyama F, Kurosumi M, et al. A multicenter phase II trial of neoadjuvant letrozole plus low-dose cyclophosphamide in postmenopausal patients with estrogen receptor-positive breast cancer (JBCRG-07) therapeutic efficacy and clinical implications of circulating endothelial cells. Cancer Med. 2018;7((6)):2442–2451. doi: 10.1002/cam4.1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ueno T, Masuda N, Kamigaki S, Morimoto T, Saji S, Imoto S, et al. Differential involvement of autophagy and apoptosis in response to chemoendocrine and endocrine therapy in breast cancer JBCRG-07TR. Int J Mol Sci. 2019;20((4)):984. doi: 10.3390/ijms20040984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Generali D, Berruti A, Cappelletti MR, Zanotti L, Brugnoli G, Forti M, et al. Effect of primary letrozole treatment on tumor expression of mTOR and HIF-1α and relation to clinical response. J Natl Cancer Inst Monogr. 2015;2015((51)):64–66. doi: 10.1093/jncimonographs/lgv018. [DOI] [PubMed] [Google Scholar]
  • 53.Bazzola L, Foroni C, Andreis D, Zanoni V, R Cappelletti M, Allevi G, et al. Combination of letrozole metronomic cyclophosphamide and sorafenib is well-tolerated and shows activity in patients with primary breast cancer. Br J Cancer. 2015;112((1)):52–60. doi: 10.1038/bjc.2014.563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Adamo B, Bellet M, Pare L, Pascual T, Vidal M, Perez Fidalgo JA, et al. Oral metronomic vinorelbine combined with endocrine therapy in hormone receptor-positive HER2-negative breast cancer SOLTI-1501 VENTANA window of opportunity trial. Breast Cancer Res. 2019;21((1)):108. doi: 10.1186/s13058-019-1195-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aurilio G, Munzone E, Botteri E, Sciandivasci A, Adamoli L, Minchella I, et al. Oral metronomic cyclophosphamide and methotrexate plus fulvestrant in advanced breast cancer patients a mono-institutional case-cohort report. Breast J. 2012;18((5)):470–474. doi: 10.1111/j.1524-4741.2012.01278.x. [DOI] [PubMed] [Google Scholar]
  • 56.Schwartzberg LS, Wang G, Somer BG, Blakely LJ, Wheeler BM, Walker MS, et al. Phase II trial of fulvestrant with metronomic capecitabine for postmenopausal women with hormone receptor-positive HER2-negative metastatic breast cancer. Clin Breast Cancer. 2014;14((1)):13–19. doi: 10.1016/j.clbc.2013.09.003. [DOI] [PubMed] [Google Scholar]
  • 57.Smith IE, Dowsett M. Aromatase inhibitors in breast cancer. N Engl J Med. 2003;348((24)):2431–2442. doi: 10.1056/NEJMra023246. [DOI] [PubMed] [Google Scholar]
  • 58.Johnston SRD, Martin LA, Leary A, Head J, Dowsett M. Clinical strategies for rationale combinations of aromatase inhibitors with novel therapies for breast cancer. J Steroid Biochem Mol Biol. 2007;106((1–5)):180–186. doi: 10.1016/j.jsbmb.2007.05.019. [DOI] [PubMed] [Google Scholar]
  • 59.Gligorov J, Azria D, Namer M, Khayat D, Spano JP. Novel therapeutic strategies combining antihormonal and biological targeted therapies in breast cancer focus on clinical trials and perspectives. Crit Rev Oncol Hematol. 2007;64((2)):115–128. doi: 10.1016/j.critrevonc.2007.06.010. [DOI] [PubMed] [Google Scholar]
  • 60.Howell A, Robertson JFR, Quaresma Albano J, Aschermannova A, Mauriac L, Kleeberg UR, et al. Fulvestrant formerly ICI 182 is as effective as anastrozole in postmenopausal women with advanced breast cancer progressing after prior endocrine treatment. J Clin Oncol. 2002;20((16)):3396–403. doi: 10.1200/JCO.2002.10.057. [DOI] [PubMed] [Google Scholar]
  • 61.Dellapasqua S, Bertolini F, Bagnardi V, Campagnoli E, Scarano E, Torrisi R, et al. Metronomic cyclophosphamide and capecitabine combined with bevacizumab in advanced breast cancer. J Clin Oncol. 2008;26((30)):4899–905. doi: 10.1200/JCO.2008.17.4789. [DOI] [PubMed] [Google Scholar]
  • 62.Di Desidero T, Xu P, Man S, Bocci G, Kerbel RS. Potent efficacy of metronomic topotecan and pazopanib combination therapy in preclinical models of primary or late stage metastatic triple-negative breast cancer. Oncotarget. 2015;6((40)):42396–410. doi: 10.18632/oncotarget.6377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Montagna E, Cancello G, Bagnardi V, Pastrello D, Dellapasqua S, Perri G, et al. Metronomic chemotherapy combined with bevacizumab and erlotinib in patients with metastatic HER2-negative breast cancer clinical and biological activity. Clin Breast Cancer. 2012;12((3)):207–214. doi: 10.1016/j.clbc.2012.03.008. [DOI] [PubMed] [Google Scholar]
  • 64.Anampa J, Chen A, Wright J, Patel M, Pellegrino C, Fehn K, et al. Phase I trial of veliparib a poly ADP ribose polymerase inhibitor plus metronomic cyclophosphamide in metastatic HER2-negative breast cancer. Clin Breast Cancer. 2018;18((1)):e135–e142. doi: 10.1016/j.clbc.2017.08.013. [DOI] [PubMed] [Google Scholar]
  • 65.Kummar S, Wade JL, Oza AM, Sullivan D, Chen AP, Gandara DR, et al. Randomized phase II trial of cyclophosphamide and the oral poly (ADP-ribose) polymerase inhibitor veliparib in patients with recurrent advanced triple-negative breast cancer. Invest New Drugs. 2016;34((3)):355–363. doi: 10.1007/s10637-016-0335-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chen Q, Xia R, Zheng W, Zhang L, Li P, Sun X, et al. Metronomic paclitaxel improves the efficacy of PD-1 monoclonal antibodies in breast cancer by transforming the tumor immune microenvironment. Am J Transl Res. 2020;12((2)):519–530. [PMC free article] [PubMed] [Google Scholar]
  • 67.Dickler MN, Rugo HS, Eberle CA, Brogi E, Caravelli JF, Panageas KS, et al. A phase II trial of erlotinib in combination with bevacizumab in patients with metastatic breast cancer. Clin Cancer Res. 2008;14((23)):7878–7883. doi: 10.1158/1078-0432.CCR-08-0141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kummar S, Ji J, Morgan R, Lenz HJ, Puhalla SL, Belani CP, et al. A phase I study of veliparib in combination with metronomic cyclophosphamide in adults with refractory solid tumors and lymphomas. Clin Cancer Res. 2012;18((6)):1726–1734. doi: 10.1158/1078-0432.CCR-11-2821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Haffty BG, Yang Q, Reiss M, Kearney T, Higgins SA, Weidhaas J, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24((36)):5652–5657. doi: 10.1200/JCO.2006.06.5664. [DOI] [PubMed] [Google Scholar]
  • 70.Cimino-Mathews A, Foote JB, Emens LA. Immune targeting in breast cancer. Oncology. 2015;29((5)):375–385. [PubMed] [Google Scholar]
  • 71.García-Teijido P, Cabal ML, Fernandez IP, Perez YF. Tumor-infiltrating lymphocytes in triple negative breast cancer the future of immune targeting. Clin Med Insights Oncol. 2016;10((Suppl 1)):31–39. doi: 10.4137/CMO.S34540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Rosato RR, Davila-Gonzalez D, Choi DS, Qian W, Chen W, Kozielski AJ, et al. Evaluation of anti-PD-1-based therapy against triple-negative breast cancer patient-derived xenograft tumors engrafted in humanized mouse models. Breast Cancer Res. 2018;20((1)):108. doi: 10.1186/s13058-018-1037-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dirix LY, Takacs I, Jerusalem G, Nikolinakos P, Arkenau HT, Forero-Torres A, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Adams S, Schmid P, Rugo HS, Winer EP, Loirat D, Awada A, 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. [DOI] [PubMed] [Google Scholar]
  • 75.Adams S, Loi S, Toppmeyer D, Cescon DW, De Laurentiis M, Nanda R, et al. Pembrolizumab monotherapy for previously untreated metastatic triple-negative breast cancer cohort B of the phase II KEYNOTE-086 study. Ann Oncol. 2019;30((3)):405–411. doi: 10.1093/annonc/mdy518. [DOI] [PubMed] [Google Scholar]
  • 76.Voorwerk L, Slagter M, Horlings HM, Sikorska K, van de Vijver KK, de Maaker M, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade the TONIC trial. Nat Med. 2019;25((6)):920–928. doi: 10.1038/s41591-019-0432-4. [DOI] [PubMed] [Google Scholar]
  • 77.Chue BMF, La Course. BD. Case report of long-term survival with metastatic triple-negative breast carcinoma treatment possibilities for metastatic disease. Medicine. 2019;98((16)):e15302. doi: 10.1097/MD.0000000000015302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chue BMF, La Course BD. Can we cure stage IV triple-negative breast carcinoma? Another case report of long-term survival (7 years) Medicine. 2019;98((38)):e17251. doi: 10.1097/MD.0000000000017251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Fuentes D, Avellanet J, Garcia A, Iglesias N, Gabri MR, Alonso DF, et al. Combined therapeutic effect of a monoclonal anti-idiotype tumor vaccine against NeuGc-containing gangliosides with chemotherapy in a breast carcinoma model. Breast Cancer Res Treat. 2010;120((2)):379–389. doi: 10.1007/s10549-009-0399-9. [DOI] [PubMed] [Google Scholar]
  • 80.Soriano JL, Batista N, Santiesteban E, Lima M, Gonzalez J, Garcia R, et al. Metronomic cyclophosphamide and methotrexate chemotherapy combined with 1E10 anti-idiotype vaccine in metastatic breast cancer. Int J Breast Cancer. 2011;2011:710292. doi: 10.4061/2011/710292. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Oncology Research and Treatment are provided here courtesy of Karger Publishers

RESOURCES