Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2020 Nov 18;157:103148. doi: 10.1016/j.critrevonc.2020.103148

De-escalating cancer treatments during COVID 19 pandemic: Is metronomic chemotherapy a reasonable option?

Palma Fedele a,*, Valeria Sanna b, Alessandro Fancellu c, Antonella Marino d, Nicola Calvani d, Saverio Cinieri d
PMCID: PMC7672334  PMID: 33254036

Graphical abstract

graphic file with name ga1_lrg.jpg

Keywords: Metronomic chemotherapy, Cancer treatment, COVID 19, De-escalation, Safety

Highlights

  • COVID 19 pandemic represents an emergency for public health services.

  • The reorganization of the healthcare system has had an important impact on the management of cancer patients.

  • Oral treatments and de-escalation strategies are encouraged.

  • Oral metronomic chemotherapy could be a reasonable treatment option in some cancer patients subgroups during COVID 19 pandemic.

Abstract

COVID 19 pandemic represents an emergency for public health services and containment measures to reduce the risk of infection have been promptly activated worldwide.

The healthcare systems reorganization has had a major impact on the management of cancer patients who are considered at high risk of infection.

Recommendations and guidelines on how to manage cancer patients during COVID 19 pandemic have been published. Oral administration of chemotherapy is recommended to limit the access of cancer patients to hospital facilities and in some cases to guarantee the continuum of care.

Low-dose metronomic administration of chemotherapy with different drugs and schedules has emerged in the last years as a possible alternative to conventional chemotherapy, due to its promising tumor control rates and excellent safety profiles. Moreover, given that many metronomic schedules use the oral route administration, it could represent a therapeutic strategy to ensure continuum of cancer care during COVID 19 pandemic.

In this review we have selected all the clinical studies that have used the metronomic strategy, especially with oral drugs, in order to identify the subgroups of cancer patients who can benefit most from a metronomic approach even during COVID 19 pandemic.

1. Introduction

The coronavirus disease 2019 (COVID-19) pandemic has forced healthcare systems to reorganize all the activities with the purpose of containing the virus infection. Medical resources have been concentrated on emergency departments and intensive care units while scheduled and non-urgent medical services have been suspended.

The reorganization of the healthcare system has had an important impact on the management of cancer patients.

Cancer patients are considered at high risk of developing coronavirus infection and its severe complications, because of their illness and immunosuppressed status (Liang et al., 2020).

In those countries where the spread of the pandemic is massive, specific measures have been taken to reduce access of cancer patients to hospitals. Elective surgeries, follow-up appointments and some types of cancer treatments have been canceled or postponed to prioritize hospital beds and care for those who are seriously ill with COVID-19 (Wang et al., 2020).

Consequently, medical oncologists must perform individual risk-benefit assessments in cancer patients before making any decision. For those patients who do not have an urgent need to start anticancer therapy, the treatment will be postponed. When the benefits for patients to undergo anti-cancer treatment outweigh the risks of being potentially exposed to the virus while traveling from home to the hospital and back, a new therapy will be initiated and in some cases the continuum of cancer care will be guaranteed.

In this regard, recommendations and practical suggestions on how to implement cancer care have been published to guide medical oncologists in the difficult decision of prioritizing patients for cancer treatments (Ontario Health and Cancer Care Ontario, 2020, NICE, 2020, Lambertini et al., 2020, You et al., 2020).

Since a priority of oncologists at this time is to minimize infection risks for cancer patients and encourage oral treatments to limit patients access to hospital facilities, we suggest to consider metronomic administration of chemotherapy as a reasonable therapeutic option to de-escalate cancer treatments and to ensure the continuum of care of some cancer patients subgroups.

Metronomic chemotherapy could allow the possibility of prolonged treatment with less side effects. It can allow the management of cancer patients at home and limit patients’ dependence on hospitals and the possibility of infection in the hospital environment.

Herein we analyze results from clinical trials that have evaluated the safety and efficacy of oral metronomic therapy in cancer patients, in order to identify the subgroups of cancer patients who are ideal candidates for metronomic chemotherapy.

2. Metronomic chemotherapy: classical and new mechanisms of action

Metronomic chemotherapy (MC) is characterized by chronic administration of chemotherapy at low doses, with a frequent schedule of administration, at close and regular intervals and with no extended interruption. MC exerts both direct and indirect effects on tumor cells and on their microenvironment and causes less severe side effects than standard chemotherapy (Hanahan et al., 2000, Romiti et al., 2017, Shitara and Nishikawa, 2018, Tanaka et al., 2009, Vincent et al., 2010).

3. Metronomic chemotherapy in breast cancer

3.1. Metastatic breast cancer

The earlier studies on MC were conducted in metastatic breast cancer (MBC) patients. Given that goals of care in MBC are to optimize both length and quality of life, MC strategies are attractive for their safe toxicity profile and good tumor control. Moreover, MC is delivered by the oral route that limits patients access to hospitals and represents the favourite route of chemotherapy administration for cancer patients (Eek et al., 2016).

The first clinical reports on MC examined the all oral combination of daily low dose of CM (cyclophosphamide 50 mg daily and methotrexate 2 days a week for a total dose of 10 mg a week) in pretreated or untreated MBC patients (Colleoni et al., 2002, Colleoni et al., 2006, Salem et al., 2008).

Subsequent studies have explored the efficacy and safety of the combination of CM with different other therapies (endocrine, anti Her2, targeted agents). The study by Aurilio et al. evaluated the combination of CM plus fulvestrant 250 mg via i.m. injection q28 days. CBR was 56% (95% CI 38–74%) and the treatment did not determine relevant toxicities (Aurilio et al., 2012).

In a small study, low-dose, oral CM combined with trastuzumab have shown substantial efficacy in metastatic HER-2 positive breast cancer and provided disease control in a significant proportion of patients (Orlando et al., 2006).

The antiangiogenic agents bevacizumab and vandetanib were combined to CM in pretreated MBC patients. The CBR was 64% in the first study that explored the efficacy of CM plus vandetanib combination in patients with anthracycline- and taxane-refractory breast cancer and reported mild side effects (Garcia-Saenz et al., 2008). The phase I study by Mayer et al. that evaluated the combination of vandetanib and CM in MBC reported mild toxicities included nausea, vomiting, fatigue and rash. Out of 20 response-evaluable patients, 10% experienced partial response and 15% stable disease ≥24 weeks (Mayer et al., 2012).

The most widely studied metronomic therapy in MBC is capecitabine-based chemotherapy.

Metronomic capecitabine (MeC) (1500 mg once daily) has shown a CBR of 62% in pretreated MBC patients and excellent safety profile, being severe toxicity rare and in all cases non-hematological (Fedele et al., 2012).

In the phase II randomized study by Stockler et al., 323 patients with MBC received one of three regimens: standard capecitabine (1000 mg/m2 twice daily for 14 of every 21 days), continuous MeC (650 mg/m2 twice daily without breaks) and classical Bonadonna CMF regimen.

Capecitabine improved overall survival and was similarly active, less toxic and more tolerable than CMF. No significant differences were observed between standard and MeC in terms of survival, tumor response and toxicity (Stockler et al., 2011).

The combination of MeC with different chemotherapeutic agents, endocrine and biological therapies have been extensively investigated in phase II and III clinical trials.

The all oral combination of MeC and cyclophosphamide was safe and effective as first or second line treatment in HER2-negative MBC patients (Yoshimoto et al., 2012, Wang et al., 2012)

The VICTOR-1 and 2 studies have investigated the all-oral metronomic combination of vinorelbine 40 mg three times a week and capecitabine 500 mg three times a day in first or subsequent lines of treatment. The metronomic schedule reported a CBR of 45.7% (95% CI 28.8–63.4) and 51.1% (95% CI 35.8–66.3) in first- and ≥ second-line therapy, respectively. The median duration of response was 11.3 and 6.4 months and PFS rates at 1 year were 24.3 and 22.2%, respectively. In triple-negative breast cancer patients (N = 28, 35%) a lower, but clinically relevant CBR (35.7; 95% CI 18.6–55.9) was observed. Side effects were: non-febrile neutropenia in 1.1%, hand-foot syndrome in 1.0%, nausea and vomiting in 1.0%, leucopenia in 0.8%, fatigue in 0.7%, and diarrhea in 0.4% (Cazzaniga et al., 2014, Cazzaniga et al., 2016).

MeC was also tested in triple-drug chemotherapy combinations.

In the phase II VEX trial the triple combination of metronomic oral vinorelbine 40 mg orally 3 times a week plus cyclophosphamide 50 mg daily and capecitabine 500 mg 3 times a day were explored in untreated metastatic triple-negative breast cancer patients. Median TTP was 6.4 months in 22/25 evaluable patients.

The combination was well tolerated: most common grade 1–2 toxicities were nausea, diarrhea, leuko-/neutropenia and reversible liver enzyme alteration. Grade ≥ 3 adverse events were uncommon (Montagna et al., 2018).

The multi-center, randomized phase II trial METEORA II is now investigating the metronomic regimen of cyclophosphamide 50 mg orally once daily continuously, capecitabine 500 mg, orally 3 times a day (1500 mg/day) continuously, vinorelbine 40 mg orally days 1, 3, 5 each week continuously, versus the conventional paclitaxel monotherapy 90 mg/m2 days 1, 8, 15 q4w as first-line or second-line treatment in patients with ER-positive/HER2-negative advanced or metastatic breast cancer (METEORA-II, 2020).

The phase II trial by Schwartzberg et al. has investigated efficacy and toxicity of MeC (1500 mg or 2000 mg daily, depending on the patient's weight) plus fulvestrant (loading dose 500 mg on day 1, 250 mg on days 15 and 29 followed by 250 mg every 28 days) in estrogen and/or progesterone receptor-positive, HER2-negative MBC, previously untreated or with ≤1 previous hormonal treatment. Median PFS was 14.98 months and median TTP was 26.94 months. Treatment was well tolerated and the most frequent adverse events were palmar-plantar erythrodysesthesia, fatigue, and nausea (Schwartzberg et al., 2014).

In the phase II trial by Dellapasqua et al. the triple combination of MeC 500 mg thrice daily plus cyclophosphamide 50 mg daily plus bevacizumab 10 mg/kg every 2 weeks has shown a high CBR in 46 untreated breast cancer patients (68% (95% CI, 51–81%). The combination resulted minimally toxic being grade 3 or 4 non hematologic adverse effects: hypertension (n = 8), transaminitis (n = 2), and nausea/vomiting (n = 2) (Dellapasqua et al., 2008).

The phase II trial that investigated the combination of MeC 500 mg thrice daily plus cyclophosphamide 50 mg daily plus bevacizumab 15 mg/kg every 3 weeks and erlotinib 100 mg daily as first line treatment in HER2-negative, hormone receptor poor MBC reported a CBR of 75% (95% confidence interval [CI], 53–90%). Median time to progression was 43 weeks (95% CI, 21–69). Toxicity was generally mild and grade 3 toxicity was rare: diarrhea (n = 1), thrombosis (n = 1), and hypertension (n = 2) (Montagna et al., 2012).

The multicenter, randomized phase III trial SAKK 24/09 compared bevacizumab with either paclitaxel or daily oral capecitabine 500 mg thrice daily plus and cyclophosphamide 50 mg daily as first-line treatment in patients with HER2-negative advanced breast cancer. No significant differences between treatment arms were reported in PFS that was 10.3 months (95% CI 8.7–11.3) in the paclitaxel arm and 8.5 months in the metronomic arm (95% CI 6.5–11.9). Less hair loss and numbness in metronomic arm were the only clinically and statistically significant differences (Rochlitz et al., 2016).

More recently a phase II trial has explored a new metronomic regimen with cyclophosphamide 50 mg daily plus capecitabine 500 mg three times a day continuously in combination with trastuzumab in 60 HER-2 positive untreated MBC. The objective response rate that was the primary endpoint of the study was 56.7% (95% CI, 44.1–68.4%) and CBR was 78.2%. Grade 3 and 4 toxicities were rare and the most commonly reported toxicities were G1 events (Orlando et al., 2017).

Metronomic therapy with oral vinorelbine has been explored in some MBC patient subgroups.

Two phase II trials have demonstrated safety and activity of oral vinorelbine metronomic (MeV) monotherapy in elderly patients with MBC (Addeo et al., 2010, De Iuliis et al., 2015).

To validate the role of MeV in the treatment of MBC patients the Name trial, a prospective randomized phase II, multicentre study, is now comparing classical treatment of i.v Vinorelbine (60 mg/m2 day 1, day 8 and day 15, every three weeks for the first cycle, hereafter 80 mg/m2 day 1 and day 8, every three weeks for the following cycles) versus MeV at daily doses of 20 or 30 mg, depending on patients’ age and body surface (Langkjer et al., 2019).

Similarly, the ongoing TEMPO- Breast 01 trial is enrolling HR positive, HER2 negative MBC patients to first-line chemotherapy with MeV 60 mg/mq per week or MeV 50 mg total dose three times per week (De la Haba et al., 2015).

3.2. Early breast cancer

In early breast cancer (EBC) the evidences currently reported show that the metronomic approach should be reserved for selected patients subgroups, such as triple negative, and as maintenance treatment.

The first study that has explored efficacy and safety of oral MC is a randomized phase III trial that compared adjuvant tegafur/uracil (UFT) to classical CMF in node negative, high risk EBC patients. Survival results were similar in both arms, but the two different schedules differed in toxicity profiles. The quality of life scores were better for patients given UFT than those given CMF (Watanabe et al., 2009).

The open-label phase III trial, IBCSG 22–00 randomized 1081 patients with ER and PGR negative EBC and any nodal status who have completed adjuvant chemotherapy to CM maintenance (cyclophosphamide 50 mg/day continuously and methotrexate 2.5 mg twice/day on days 1 and 2 of every week for 1 year) or to no CM.

The metronomic CM maintenance therapy did not produce a significant reduction in DFS that was the primary endpoint of the study (DFS at 5 years was 78.1% in the CM group vs 74.7% in the no CM group (hazard ratio [HR] = 0.84, p = 0.14). There was a non-statistically significant reduced HR (n = 340; HR, 0.72; 95% CI, 0.49–1.05) in the triple-negative, node-positive subgroup. Moreover, the CM maintenance chemotherapy was associated to grade 3 or 4 treatment-related adverse events that occurred in 14%. The most common side effect was elevated serum transaminases (7%) and leukopenia (2%). Two patients in the cyclophosphamide/methotrexate group developed acute myeloid leukemia (Colleoni et al., 2016).

Metronomic CM (cyclophosphamide 50 mg daily: methotrexate 2.5 mg BID on days 1, 2 of each week) was administered for 1 year after adjuvant therapy completion to patients with TNBC to improve their DFS and OS in a randomized phase III study by Nasr et al. The authors reported significantly better OS for those TNBC patients who received CM maintenance chemotherapy after adjuvant carboplatin versus patients who did not (Nasr et al., 2015).

MeC was administered to EBC patients as maintenance after adjuvant chemotherapy in two phase II trials that confirmed efficacy and good tolerability of the extended metronomic approach (Shawky and Galal, 2014, Alagizy et al., 2015).

Results from ongoing randomized adjuvant trials (ABCDE trial and MACRO trial) will better address the role of metronomic chemotherapy with CM or capecitabine in the maintenance treatment of EBC (Mayer et al., 2016, MACRO, 2020)

4. Metronomic chemotherapy in colorectal cancer

In preclinical studies MeC for colon cancer xenografts and colon cancer cells has proved to inhibit angiogenesis, decrease VEGF and microvessel density and increase antiangiogenic protein thrombospondin-1 (TSP-1) (Shi et al., 2014).

The best clinical experience in colorectal cancer (CRC) derives from advanced disease where lowered but prolonged doses of standard chemotherapy have been used especially with the aim of targeting angiogenesis. The chemotherapeutic agents mostly studied for a metronomic approach in CRC are the fluoropyrimidines, predominantly capecitabine. Other agents evaluated in very few phase II studies are: tegafur/uracil (UFT), irinotecan, cyclophosphamide.

The role of MeC has been particularly evaluated in phase II studies in the palliative setting of advanced pretreated patients and in one phase III trial in the maintenance therapy setting.

In 2000 a randomized phase II trial compared 3 capecitabine schedules; arm A: 1331 mg/m2/day continuous dosing, arm B: 2510 mg/m2/day [2 weeks on, 1 week off] and arm C: an additional leucovorin-containing arm [capecitabine 1657 mg/m2/day plus leucovorin 60 mg/day, 2 weeks on and 1 week off]. Time to progression was longer in the group that received the intermittent capecitabine dose of 2510 mg/m2/day (arm A 127 days, arm B 230 days, and arm C 165 days) and the response rates were similar in the 3 arms (arm A 21%, arm B 24%, and arm C 23%) (Van Cutsem et al., 2000).

In one retrospective study the patients who received a continuous fixed dose of capecitabine 1500 or 2000 mg daily had low toxicity profiles and no patients who were treated with capecitabine as a single agent had side effects of any grade (Lokich, 2004).

Continuous administration of a fixed daily dose of capecitabine was effective and well tolerated with a low toxicity profile (Lokich, 2004, Budman et al., 1998).

One phase 3 randomized controlled trial (CAIRO 3 study) was planned to ascertain the efficacy of maintenance chemotherapy with MeC plus bevacizumab after an induction treatment with six 3-week cycles of capecitabine, oxaliplatin and bevacizumab (CAPOX-B). 558 mCRC patients were randomized into either the maintenance or the observation group on a 1:1 basis. Capecitabine 625 mg/m2 orally twice a day for 3 weeks and bevacizumab 7.5 mg/m2 intravenously every 3 weeks was the maintenance treatment. With a median follow-up of 48 months PFS was significantly longer in the maintenance group (8.5 months vs 11.7 months). Furthermore, the incidence of chemotherapy-related leukopenia, peripheral neurotoxicity and other serious toxic reactions was only increased by 5–10% in the maintenance group compared with the observation group which was completely tolerated by patients and thus MeC chemotherapy combined with bevacizumab proved to be an effective and low-toxic maintenance therapy (Simkens et al., 2015).

On the contrary, a different study, the Italian phase II, no-profit, multicenter MOMA trial did not show the same positive results. 232 patients with unresectable mCRC were randomized to receive up to 8 cycles of FOLFOXIRI plus bevacizumab followed by bevacizumab (arm A) or the same induction regimen followed by bevacizumab plus MeC (capecitabine 500 mg three times per day and cyclophosphamide 50 mg orally daily arm B) until disease progression. The primary endpoint was PFS. At a median follow-up of 47.8 months, 210 and 164 progression and death events were registered. Median PFS was 10.3 and 9.4 months in arm B and A, respectively (HR: 0.94 [70% confidence interval {CI}: 0.82 and 1.09], p: 0.680). No significant differences were reported in terms of overall survival (OS) (median OS arm B/A: 22.5/28 months; HR: 1.16 [95% CI: 0.99 and 1.37], p: 0.336). The authors concluded that the addition of MeC to maintenance with bevacizumab did not improve significantly PFS of mCRC patients (Cremolini et al., 2019). Recently Shi et al. published a project intended to study the efficacy and safety of MeC in the maintenance treatment of advanced CRC. The study is a prospective, randomized, open label, phase II clinical trial in which patients with mCRC, who had responded well after 16–18 weeks of standard doublet chemotherapy as induction therapy, were randomly assigned to the MeC group (capecitabine 500 mg twice a day orally) and capecitabine conventional chemotherapy (capecitabine 1000 mg/m2 twice a day orally, d1–14, 3 times weekly (q3w). The aim of the study is to demonstrate that MeC is non-inferior to capecitabine conventional chemotherapy as maintenance treatment in patients who have responded to 16–18 weeks first-line chemotherapy in mCRC. The duration of disease control after randomization and progression-free survival after enrollment are the primary endpoints. Overall survival, safety, and quality of life are the secondary endpoints. The sample size required to achieve the research objectives of this project is 79 patients in each group. The study enrollment started on 29 January 2018 and will last for 36 months. After the start of the study, the first 30 months have consisted of inclusion and follow up of patients. The last 6 months consisted of follow up and analysis of results. The study will end on 29 January 2021. This project is intended to explore the strategy of low toxicity, high efficiency, economy and individualization of MeC in the maintenance treatment of advanced colorectal cancer which is suitable for China's national conditions and pharmacoeconomics. It has great prospects for clinical application and a clear socioeconomic value (Shi et al., 2020).

Metronomic regimens could be an inviting option also for frail mCRC patients. With continuous, low-dose administration of capecitabine (500 mg twice or three times a day) elderly or heavily pretreated patients with mCRC showed good disease control and minimal toxicity without impairment to quality of life. A study by Romiti et al. retrospectively evaluated the activity and safety of MeC at the dose of 1500 mg daily in 86 frail patients. Overall disease control rate was 26% with a 2% partial response and 23% stable disease. Nineteen percent of patients were progression-free for 6 months and the median OS was 8 months. No grade 4 toxicity was observed (Romiti et al., 2015).

A different trial in pretreated frail elderly patients with mCRC evaluated the efficacy and toxicity profile of MeC (1000 mg twice daily), oxaliplatin (65 mg/m2) and bevacizumab (7.5 mg/m2). Median progression-free survival was 12.3 months with 86.7% reaching six months. No grade 4 toxicity was observed (Carreca et al., 2011).

A different study retrospectively evaluating MeC (1500 mg daily) in mCRC patients reported a median TTP of 6.3 months and a tolerable toxicity profile (Borgonovo et al., 2016).

Based on these data metronomic chemotherapy, especially capecitabine, should probably be taken into major consideration as a reasonable and feasible option in a further line of therapy in pretreated mCRC pts and/or frail/elderly pts during the COVID-19 pandemic.

5. Metronomic chemotherapy in prostate cancer

In castration-resistant prostate cancer (CRPC), despite the availability of new anti-testosterone drugs, a debate still exists on the optimal treatment, especially because most patients are elderly and frail.

Recent reports highlight the role of MC for those patients who progressed on standard therapy, as well as docetaxel-resistant patients. In fact, many patients in this setting could be unfit for conventional treatment (Van Dodewaard-de Jong et al., 2015).

The main part of the studies investigating MC in CRPC was on the effects of cyclophosphamide alone (Caffo et al., 2019) or combined in pretreated patients.

Metronomic cyclophosphamide was well tolerated and showed efficacy also in hormone naïve patients (Calcagno et al., 2016).

Combinations of cyclophosphamide plus steroids were effective and safe in pretreated patients with no febrile neutropenia and beneficial effects in 50–79% of patients were reported, including reduction of PSA levels (Glode et al., 2003, Ladoire et al., 2010, Calvan iN et al., 2019).

Fea et al. analyzed the pharmacologic toxicity of metronomic oral cyclophosphamide in a group of heavily pretreated patients and did not find any grade 3 or 4 toxicity. Thus, in their cohort none of the patients discontinued therapy because of toxicity. The most common adverse events were asthenia G1, anemia G1-2 and leukopenia G1 (Fea et al., 2016).

Metronomic cyclophosphamide was well tolerated also in those studies that evaluated the combination with iv chemotherapy. In a study including 41 patients, where cyclophosphamide was combined with docetaxel and prednisone, no grade 4 toxicities were reported, while grade 3 neutropenia was 5%; thrombocitopenia, stomatitis and diarrhea were 2.5%. These side effects were related to docetaxel treatment. Neither major cardiovascular events nor toxicity-related deaths were observed (Derosa et al., 2014).

Only in one study, a moderate rate of myelotoxicity (about 12%) was reported in a cohort of patients with extensive bone metastasis (Jeong and Lee, 2017).

Other effective combination regimens (for example with corticosteroids, diethylstilbestrol or colecoxib and methotrexate) were studied in pretreated patients with similar results (lowering PSA; good tolerance) (Hellerstedt et al., 2003, Muraki et al., 2012, Khan et al., 2011).

In a phase II trial, in which cyclophosphamide 50 mg daily was combined with methotrexate in pretreated patients, PSA lowering was observed in 25% of patients (Gebbia et al., 2011).

In a different study of docetaxel naive patients, a 50% of reduction of PSA was observed with use of cyclophosphamide and estramustine. The safety profile was considered good, without G3/4 toxicity (Bracarda et al., 2000).

As shown in Table 1 all the studies that evaluated MC alone or in combinations in pretreated or naïve prostate cancer patients have demonstrated a manageable toxicity profile.

Table 1.

Main toxicities reported with metronomic chemotherapy in patients with prostate cancer.

Author Year No. of patients Drug FN
N
SAE
N (worst event)
Maulard Durdux (Maulard-Durdux et al., 1996) 1996 20 CP-E 0 0
Bracarda (Bracarda et al., 2000) 2000 32 CP-Estra 0 0
Nishimura (Nishimura et al., 2001) 2001 21 CP-Estra-U-T 0 d.n.r (mild toxicity/well tolerated)
Glode (Glode et al., 2003) 2003 34 CP-DEX 0 d.n.r (mild toxicity/well tolerated)
Robles (Robles et al., 2003) 2003 14 V-PD 0 d.n.r (mild toxicity/well tolerated)
Hellerstedt (Hellerstedt et al., 2003) 2003 36 CP-PD-DE 0 d.n.r (mild toxicity/well tolerated)
Lord (Lord et al., 2007) 2007 58 CP 0 0
Fontana (Fontana et al., 2009) 2009 28 CP-Cel-DEX 0 0
Nelius (Nelius et al., 2010) 2010 17 CP 0 0
Ladoire (Ladoire et al., 2010) 2010 23 CP-PL 0 d.n.r (mild toxicity/well tolerated)
Gebbia (Gebbia et al., 2011) 2011 58 CP-MTX 0 0
Jellvert (Jellvert et al., 2011) 2011 17 CP-Estra-E-K 0 0
Hatano (Hatano et al., 2011) 2011 57 CP-U-T-DEX 0 1 (neutropenia)
Meng (Meng et al., 2012) 2012 28 CP-Tha-Cap 0 0
Yashi (Yashi et al., 2014) 2014 14 CP 0 0
Derosa (Derosa et al., 2014) 2014 41 CP-Doc 0 0
Barroso-Sousa (Barroso-Sousa et al., 2015) 2015 40 CP-PD 0 0
Fea (Fea et al., 2016) 2016 12 CP 0 0
Di Desidero (Di Desidero et al., 2016) 2016 41 V-DEX 0 0
Tralongo (Tralongo et al., 2016) 2016 26 V 0 0
Calcagno (Calcagno et al., 2016) 2016 38 CP 0 0
Jeong (Jeong and Lee, 2017) 2017 60 CP-DEX-Cel 0 6 (myelophthisic anemia)
Dabkara (Dabkara et al., 2018) 2018 18 CP-PL 0 0
Caffo (Caffo et al., 2019) 2019 74 CP 0 1 (non neutropenic infection)
Calvani (Calvan iN et al., 2019) 2019 37 CP 0 0

(CP: cyclophosphamide; Estra: estramustina; L: lenalinomide; V: vinorelbine; E: etoposide; U: uracil; T: tegafur; DEX: dexamethasone; PD: prednisone; PL: prednosolone; DE: diethylstilbestrol; MTX: methotrexate; cel: celecoxib; Tha: thalidomide; Cap: capecitabine; Doc: docetaxel; K: ketoconazole; d.n.r: details not reported; FN: febrile neutropenia; SAE: severe adverse event, defined as any >3 grade toxicity or treatment interrupted.

6. Metronomic chemotherapy in kidney cancer

Kidney cancer is usually thought to be resistant to standard chemotherapy and even targeted drugs seem to be only temporarily effective because of resistance.

As target therapy and immunotherapy have improved the prognosis of metastatic renal cancer, the issues of quality of life and of pharmacologic tolerance are of paramount importance in this scenario.

In particular, the resistance to these drugs still remains a problem. Further treatment strategy and MC could be an opportunity for renal cancer cure.

Few studies have addressed the role of MC in patients with metastatic renal cancer. In 2010 Bellmunt reported a clinical benefit in 87% of cases and no G3/4 hematological toxicity in a group of 44 patients taking MeC (Bellmunt et al., 2010)

In a pretreated population, a combination of capecitabine and antinflammatory politherapy (pioglitazone, IFN, etorocoxib) was shown to have a response rate of 35%. No febrile neutropenia and skin toxicity was seen (Walter et al., 2012).

A phase II trial confirmed a low grade of toxicity with cyclophosphamide and a long clinical benefit (24 weeks] in 40% of patients (Tupikowski et al., 2015).

There is evidence of a synergistic activity of target therapy associated with metronomic chemotherapy. There is direct effect of pazopanib on renal cancer cells, resulting in increased intracellular concentration of topotecan (Jedeszko et al., 2015). Although these data are encouraging, further studies are needed to make this strategy applicable on a large scale.

Certainly, the manageability and low toxicity profile make MC particularly attractive also in kidney cancer.

Table 2 reports safety details of the more significant phase II trials in kidney cancer.

Table 2.

Main toxicities reported with metronomic chemotherapy in patients with kidney cancer.

Author Year No. of patient Drug FN
N
SAE
N (worst event)
Bellmunt (Tupikowski et al., 2015) 2010 44 Cape-Gem-Sor 0 1 (PE)
Walter (Jedeszko et al., 2015) 2012 45 Cape-IFN–Pi_Eto 0 16 (Hand and foot Syndrome)
Tupikowsky (Kamat et al., 2007) 2015 30 IFN-CP 0 d.n.r (mild toxicity/well tolerated)

T: tegafur; Cape: capecitabina; Gem: gemcitabine; Sor: sorafenib; IFN: interferon; Pi: pioglitazone; Eto: etoricoxib CP: cyclophosphamide; T: topotecan; Pa: pazopanib; d.n.r: details not reported; FN: febrile neutropenia; SAE: severe adverse event, defined as any >3 grade toxicity or treatment interrupted; PE: pulmonary embolism.

7. Metronomic chemotherapy in ovarian cancer

In ovarian cancer high level of response rate can be reached with debulking surgery and/or platinum based chemotherapy. However, relapse still occurs.

In ovarian cancer, angiogenesis plays an important role, thus MC could be an interesting opportunity (Kamat et al., 2007).

In most studies on ovarian cancer, MC has been used in relapsed/refractory ovarian carcinoma or in combination with standard chemotherapy to improve outcomes due to antiangiogenic effect with minimal toxicity.

This frail population, generally heavily pretreated or unfit for iv chemotherapy because of complication of surgery, could have beneficial effects by continuous administration of low doses of chemotherapy.

In animal models metronomic dosage of oral cyclophosphamide was proven to be safe in combination with irinotecan or pazopanib, with modest lowering of white blood cells and weight loss (Hashimoto et al., 2010).

Numerous experiences of metronomic cyclophosphamide in ovarian cancer have shown an optimal safety profile with an overall survival benefit from 12 to 20 months in pretreated advanced disease (Samaritani et al., 2007, Ferrandina et al., 2014).

Combinations of metronomic cyclophosphamide with 5-fluorouracile and temozolamide have given similar results (Kerbel, 2007, Bhattacharyya et al., 2017).

MC with some antiangiogenetic agents is an interesting area for research for first-line, maintenance and salvage therapy (Sanchez-Munoz et al., 2010, Alvarez et al., 1999, Jurado et al., 2008, Chura et al., 2007).

Cyclophosphamide metronomic regimen was evaluated as maintenance therapy in a retrospective study of ovarian cancer patients after surgery or complete response to standard neoadjuvant therapy (platinum salt based). In this population MC of cyclophosphamide and methotrexate was compared to observation alone with a benefit of 3 months without any grade 3 or 4 toxicity (Pandey et al., 2016a).

Oral etoposide is used as metronomic therapy in metastastic and pretreated ovarian cancer. A low level of toxicity and an overall survival of about 16 months indicate a good potential for this therapy, especially in the setting of platinum refractory patients (Markman et al., 1992, Kucukoner et al., 2012).

Table 3 shows main toxicity reported with metronomic chemotherapy in ovarian cancer

Table 3.

Main toxicities reported with metronomic chemotherapy in patients with ovarian cancer.

Author Year No. of patient Drug FN
N
SAE
N (worst event)
Beck (Beck and Boyes, 1968) 1968 126 CP 0 d.n.r (mild toxicity/well tolerated)
Markman (Markman et al., 1992) 1992 18 E 0 2 (neutropenia)
Chura (Chura et al., 2007) 2007 15 CP-Beva 0 0
Garcia (Garcia et al., 2008) 2008 70 CP-Beva 0 2 (linfopenia)
Jurado (Jurado et al., 2008) 2008 9 CP-Beva 0 0
Ferrandina (Ferrandina et al., 2014) 2014 54 CP 0 0
Handolias (Handolias et al., 2016) 2016 23 CP 0 2 (non-hematological)
Wong (Wong and Liu, 2017) 2017 20 CP 0 0
Sharma (Sharma et al., 2019) 2019 36 Pa-CP 0 d.n.r (mild toxicity/well tolerated), 5 G3/4 mucositiis

CP: cyclophosphamide; E: etoposide; Pa: pazopanib; beva: Bevacizumab; d.n.r: details not reported; FN: febrile neutropenia; SAE: severe adverse event, defined as any >3 grade toxicity or treatment interrupted.

8. Metronomic chemotherapy in lung cancer

Although the introduction of immunotherapy has changed the prognosis of lung cancer, it remains a big killer. Most research efforts in the management of non-small cell lung cancer (NCSLC) patients are focused on discovering agents and combinations of agents, doses and dose schedules that maximally kill tumor cells while minimizing the toxicity to the host, especially beyond the second line.

MC has been used for patients who are ineligible for standard treatment options. MC is a manageable therapy in frail patients with lung cancer with low percentage of severe toxicity, also beyond second line treatments (Kontopodis et al., 2013).

For NSCLC metronomic regimens were tested in first line treatment and for maintenance or salvage therapy. Recently, a well conducted metanalysis has been published about the use of MeV with demonstration of a good benefit-risk ratio. The safety profile of oral vinorelbine appears to be better than iv regimen (Pujol et al., 2019).

The overall survival with front line vinorelbine was 7–12 months. The best results were in young patients and in patients with a good PS (Bilir et al., 2017, Camerini et al., 2015, Katsaounis et al., 2015).

The combination vinorelbine with sorafenib resulted in overall survival of about 8 months (Tan et al., 2015).

In SCLC patients oral etoposide is the most experienced drug for salvage therapies but gives a low overall survival (about 4 months) compared with iv therapy (Pfeiffer et al., 1997).

Etoposide was used also in NSCLC with an OS of 9 months and a good safety profile (Kakolyris et al., 1998, Surmont et al., 2009).

In combination with bevacizumab and cisplatin, oral etoposide showed an overall response rate of 45.2%, without G4 hematological toxicity (Correale et al., 2006).

The same combination administered as maintenance therapy reached an overall survival of 13.2 months (Petrioli et al., 2015).

Because of the high incidence of brain metastases in patients with lung cancer, many studies have evaluated the role of MC in combination with radiotherapy (RT) to improve tolerability. Temozolamide was studied as MC in advanced stage NSCLC with brain metastases. The low dose temozolamide schedule reported a response rate of about 50% in association with RT (Addeo et al., 2008).

The combination of metronomic etoposide to cisplatin and bevacizumab was safe and effective during RT (Pastina et al., 2017).

Metronomic cyclophospamide plus RT shows a significantly higher PFS clinical benefit as radiosensitizer in NCSLC frail patients (Revannasiddaiah et al., 2015). Further histological sub-group analysis demonstrated that there was an enhanced outcome with the addition of metronomic cyclophosphamide to RT for patients with adenocarcinoma histology (3.5 vs. 2.4 months; p = 0.0053), but there was no benefit for patients with squamous cell histology (2.6 vs. 2.6 months; p = 1).

Table 4 shows main toxicities reported with metronomic chemotherapy in lung cancer.

Table 4.

Main toxicities reported with metronomic chemotherapy in lung cancer.

Author Year No. of patient Drug FN
N
SAE
N (worst event)
Kakolyris (Kakolyris et al., 1998) 1998 61 E 0 d.n.r (mild toxicity/well tolerated)
Kouroussis (Kouroussis et al., 2009) 2009 31 TMZ 0 2 (lymphopenia)
Correale (Correale et al., 2006) 2011 45 E-Beva_cis 0 0
Kontopodis (Kontopodis et al., 2013) 2013 46 V 4 8
Camerini (Camerini et al., 2015) 2015 43 V 0 0
Pastina (Pastina et al., 2017) 2017 69 E-Beva-Cis-RT 0 0
Mencoboni (Mencoboni et al., 2017) 2017 76 V 1 1 (diarrhea)
Banna (Banna et al., 2017) 2018 41 V 0 0
Pasini (Pasini et al., 2018) 2018 92 V 1 4 (neutropenia)
D’Ascanio (D’Ascanio et al., 2018) 2018 44 V 0 0

E: ethoposide; V: vinorelbine; SO: sorafenib; Beva: bevacizumab; TMZ: temozolamide; Cis: Cisplatunum. d.n.r: details not reported; FN: febrile neutropenia; RT: radiotherapy; SAE: severe adverse event, defined as any >3 grade toxicity or treatment interrupted.

9. Metronomic chemotherapy in head and neck cancer

In patients with head and neck cancer (HNC) there are only limited management options due to the very frailty of this group of patients. MC may have a role for its good safety profile, as shown in Table 5 . Low-dose, continuous metronomic drugs were particularly studied in the contest of platinum-refractory patients and the commonest adverse event was fatigue (G2–G3), while no febrile neutropenia was reported (Patil et al., 2019).

Table 5.

Main toxicities reported with metronomic chemotherapy in patients with head and neck cancer.

Author Year No. of patient Drug FN
N
SAE
N (worst event)
Pai (Pai et al., 2013) 2013 32 Col-MTX 0 0
Pandey (Pandey et al., 2016b) 2016 335 Col-MTX 0 0
Patil (Patil et al., 2015) 2020 76 Erlo-Col-MTX 0 d.n.r (mild toxicity/well tolerated); G3/5 iponatremia and neutropenia

MTX: methotrexate, Col: colecoxib; Erlo: erlotinib; d.n.r: details not reported; FN: febrile neutropenia; SAE: severe adverse event, defined as any >3 grade toxicity or treatment interrupted.

MC in recurrent HNC has shown good disease-control rates with effective palliation, minimal toxicity and preserved quality of life (Noronha et al., 2016, Patil et al., 2015).

In oral cancer methotrexate per os was evaluated with celecoxib with a significant beneficial of 15%; disease free survival was 13 months (Pai et al., 2013, Pandey et al., 2016b).

10. Metronomic chemotherapy in melanoma

Although recent advances with target therapy, immunotherapy and combinations in the treatment of metastatic melanoma (MM), there is still the need for new well-tolerated therapies for patients who have resistance or have a bad tolerance to those treatments.

Besides, as the progression-free survival duration is prolonged, the risk of treatment resistance increases.

It was shown that angiogenesis has an impact in melanoma, so it can be postulated that also MC has a role in this setting (Ugurel et al., 2001).

In MM the main experience with MC is in the use of alkylating agents.

The first study of MC in MM explored the combinations of metronomic paclitaxel and celecoxib, but the toxicity was higher compared with oral regimen in other cancers and may be correlated to iv paclitaxel (Bhatt et al., 2010).

In a group of unfit elderly patients, treatment with cyclophosphamide reached an overall survival of 8 months, ranging from 4 to 37 (Borne et al., 2010)

Similar results were reported also with cyclophosphamide combined with dendritic cell vaccine and a COX-2 inhibitor (Ellebaek et al., 2012).

A different trial showed a good tolerance using temozolamide in association with cisplatin; the combination resulted in an overall survival of 50 weeks (Simeone et al., 2009). Table 6 shows main toxicities reported with metronomic chemotherapy in patients with melanoma.

Table 6.

Main toxicities reported with metronomic chemotherapy in patients with melanoma.

Author Year No. of patient Drug FN
N
SAE
N (worstevent)
Bhatt (Bhatt et al., 2010) 2010 20 Pacli-cel 0 d.n.r (mildtoxicity/welltolerated); 4 evtherapyrelatedtoxicity G3/4)
Borne(Borne et al., 2010) 2010 13 CP 0 1 linfopenia
Ellebaek (Ellebaek et al., 2012) 2012 28 CP-IL2 0 0
Simeone (Simeone et al., 2009) 2016 33 TMZ- Cis 0 0

d.n.r: detailsnotreported; FN: febrile neutropenia; SAE: severe adverse event, defined as any grade 4 toxicity or treatment interrupted.

11. Metronomic chemotherapy in brain tumors

Glioblastoma is the most common malignant brain tumor. Standard therapy for glioblastoma includes surgery, radiotherapy and temozolamide. Few therapies are approved for recurrent disease and the prognosis is very poor.

A metronomic approach for glioblastoma can be useful, because low dose chemotherapy could be well tolerated in a very frail population.

Several clinical trials have studied the use of temozolamide, etoposide and cyclophosphamide which have the advantage of being manageable also in the outpatient setting, although no benefits in survival rate have been demonstrated (Kesari et al., 2007).

Temozolamide was safe with radiotherapy and the overall survival was about 7 months (Clarke et al., 2009); moreover, metronomic temozolamide was active for those patients who are refractory to standard cyclic treatment (Kong et al., 2010, Perry et al., 2010).

The addition of bevacizumab has conflicting results. Bevacizumab used with a low dose temozolamide schedule seems to get a worse response (Omuro et al., 2013)

There are suggestions that MeC and bevacizumab could be used with success (Peereboom et al., 2019)

In 2013 Chen conducted a metanalysis comparing metronomic and standard temozolamide regimens; no statistically significant difference was found between metronomic and standard schedules for response rate and no difference for overall survival at six and 12 months were reported (Chen et al., 2013).

In association with antinflammatory colecoxib, temozolamide had a very good safety profile without any G3/4 toxicity (Stockhammer et al., 2010).

In a different study with temozolamide and bevacizumab and etoposide, only 1 episode of G4 neutropenia was seen (Reardon et al., 2009, Reardon et al., 2011).

Zustovich et al. studied another tyrosine kinase inhibitor, sorafenib, twice daily with metronomic temozolamide; 6-month PFS was 26% and median OS was 7.4 months (Zustovich et al., 2013).

Table 7 shows main toxicities reported with metronomic chemotherapy in patients with brain tumors.

Table 7.

Main toxicities reported with metronomic chemotherapy in patients with brain tumors.

Author Year No. of patient Drug FN
N
SAE
N (worst event)
Kesari (Kesari et al., 2007) 2007 48 CP-E 0 2 (costipation)
Clarke (Clarke et al., 2009) 2009 43 TMZ 0 0
Reardon (Reardon et al., 2009) 2009 59 E-Beva 0 0
Kong (Kong et al., 2010) 2010 38 TMZ 0 0
Stockhammer 2010 28 TMZ-Col 0 0
Reardon (Reardon et al., 2011) 2011 23 TMZ/E-Bev 0 1 (neutropenia)
Omuro (Omuro et al., 2013) 2013 47 TMZ 0 1 (linfopenia,thrombocitopenia)
Zustovich (Zustovich et al., 2013) 2013 43 TMZ -SO 0 d.n.r (mild toxicity/well tolerated), 5 G3/4 Hand foot syndrome)
Welzel (Welzel et al., 2015) 2015 146 TMZ EBRT Col 0 0
Peereboom (Peereboom et al., 2019) 2019 11 Bev-cape 0 d.n.r (mild toxicity/well tolerated

GB: glioblastoma; CP: cyclophosphamide, TMZ: temozolamide; E: etoposide; Beva: bebavizumab; SO: sorafenib; Col: colecoxib; d.n.r: details not reported; FN: febrile neutropenia; SAE: severe adverse event, defined as any grade 4 toxicity or treatment interrupted.

12. Conclusions

Data reported so far have shown that oral metronomic regimens could be a reasonable treatment option in cancer patients. The low toxicity profile supports the chronic administration of the treatment especially in metastatic and pretreated patients and it could ensure the continuum of cancer care, preserving the prognosis of cancer patients also during COVID 19 pandemic. Low dose metronomic chemotherapy has a favorable safety profile and this reduces the need for hospitalization of cancer patients. This is particularly useful for prioritize hospital beds and care for those who are seriously ill with COVID-19.

Moreover, it represents a treatment option for the frail elderly. The frail elderly are often excluded from clinical trials, but they represent the majority of patients at risk of both cancer development and serious complications occurrence by Covid infection, if treated with a standard dose chemotherapy. Several studies demonstrate that metronomic therapy with different drugs and schedules is safe also in elderly patients. Safety is of the outmost importance in palliative situations. In general there was no life-threatening adverse event or major risk of infection with metronomic regimens.

For combination therapy it seems that adverse events were linked to other factors not related to metronomic therapy. To date the studies on this topic present some limits, the main ones being the small sample size and the retrospective design. In addition, inclusions criteria were based on clinical evaluation: dependence level, comorbidities and number of drugs taken. Thus, at present solid evidences on the role of MC in the different neoplasms are lacking.

Nonetheless, the results are encouraging for the use of metronomic drugs in a population presenting various poor prognosis factors, such as age, comorbidities, pre-treatment lymphopenia and unfitness for usual chemotherapy. The metronomic schema seems to be a good alternative treatment, because of its safety and easy oral taking, enabling patients to stay at home longer. Further prospective randomized studies are needed to confirm more accurately the efficacy of specific metronomic regimens.

Authors’ contribution

Palma Fedele, Valeria Sanna: conceptualization, methodology, software. Palma Fedele, Valeria Sanna, Antonella Marino, Alessandro Fancellu: writing – original draft preparation. Saverio Cinieri, Nicola Calvani: visualization, supervision. Palma Fedele, Valeria Sanna: writing – reviewing and editing.

Funding

None.

Conflict of interest

The authors declare no conflict of interest related to this study. All authors declare no financial and personal relationships with other people or organizations that could inappropriately influence (bias) their work.

Acknowledgement

We thank Antonio Guadalupi for his assistance with the manuscript.

References

  1. Addeo R., DeRosa C., Faiola V. Phase 2 trial of temozolomide using protracted low-dose and whole-brain radiotherapy for non-small cell lung cancer and breast cancer patients with brain metastases. Cancer. 2008;113(9):2524–2531. doi: 10.1002/cncr.23859. [DOI] [PubMed] [Google Scholar]
  2. Addeo R., Sgambato A., Cennamo G. Low-dose metronomic oral administration of vinorelbine in the first-line treatment of elderly patients with metastatic breast cancer. Clin. Breast Cancer. 2010;10:301–306. doi: 10.3816/CBC.2010.n.039. [DOI] [PubMed] [Google Scholar]
  3. Alagizy H.A., Shehata M.A., Hashem T.A. Metronomic capecitabine as extended adjuvant chemotherapy in women with triple negative breast cancer. Hematol. Oncol. Stem Cell Ther. 2015;8:22–27. doi: 10.1016/j.hemonc.2014.11.003. [DOI] [PubMed] [Google Scholar]
  4. Alvarez A.A., Krigman H.R., Whitaker R.S. The prognostic significance of angiogenesis in epithelial ovarian carcinoma. Clin. Cancer Res. 1999;5(3):587–591. [PubMed] [Google Scholar]
  5. Aurilio G., Munzone E., Botteri E. 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]
  6. Banna G., Anile G., Castaing M. Palliative chemotherapy with oral metronomic vinorelbine in advanced non-small cell lung cancer (NSCLC) patients unsuitable for chemotherapy. J. Thorac. Oncol. 2017;12:S907–S908. [Google Scholar]
  7. Barroso-Sousa R., da Fonseca L.G., Souza K.T. Metronomic oral cyclophosphamide plus prednisone in docetaxel pretreated patients with metastatic castration-resistant prostate cancer. Med. Oncol. 2015;32:443–444. doi: 10.1007/s12032-014-0443-4. [DOI] [PubMed] [Google Scholar]
  8. Beck R.E., Boyes D.A. Treatment of 126 cases of advanced ovarian carcinoma with cyclophosphamide. Can. Med. Assoc. J. 1968;98(11):539–541. [PMC free article] [PubMed] [Google Scholar]
  9. Bellmunt J., Trigo J.M., Calvo E. Activity of a multitargeted chemo-switch regimen (sorafenib, gemcitabine, and metronomic capecitabine) in metastatic renal-cell carcinoma: a phase 2 study (SOGUG-02-06) Lancet Oncol. 2010;11(4):350–357. doi: 10.1016/S1470-2045(09)70383-3. [DOI] [PubMed] [Google Scholar]
  10. Bhatt R.S., Merchan J., Parker R. A phase 2 pilot trial of low-dose, continuous infusion, or ‘metronomic’ paclitaxel and oral celecoxib in patients with metastatic melanoma. Cancer. 2010;116(7):1751–1756. doi: 10.1002/cncr.24902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bhattacharyya G.S., Malhotra H., Parikh P.M. Phase II study of metronomic cyclophosphamide and temozolomide in platinum resistance ovarian cancer. ASCO Annual Meeting Proceedings. J. Clin. Oncol. 2017;33(15S):e16575. [Google Scholar]
  12. Bilir C., Durak S., Kizilkaya B. Efficacy of metronomic vinorelbine in elderly patients with advanced non-small-cell lung cancer and poor performance status. Curr. Oncol. 2017;24:e199–e204. doi: 10.3747/co.24.3486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Borgonovo K.F., Petrelli F., Cabiddu M. Monoinstitutional retrospective analysis of metronomic capecitabine (mc) schedule in the treatment of metastatic colorectal cancer European Society for Medical Oncology Congress Proceedings. Ann. Oncol. 2016;27(4S):D48. [Google Scholar]
  14. Borne E., Desmedt E., Duhamel A. Oral metronomic cyclophosphamide in elderly with metastatic melanoma. Investig. New Drugs. 2010;28:684–689. doi: 10.1007/s10637-009-9298-5. [DOI] [PubMed] [Google Scholar]
  15. Bracarda S., Tonato M., Rosi P. Oral estramustine and cyclophosphamide in patients with metastatic hormone refractory prostate carcinoma: a phase II study. Cancer. 2000;88:1438–1444. [PubMed] [Google Scholar]
  16. Budman D.R., Meropol N.J., Reigner B. Preliminary studies of a novel oral fluoropyrimidine carbamate: capecitabine. J. Clin. Oncol. 1998;16:1795–1802. doi: 10.1200/JCO.1998.16.5.1795. [DOI] [PubMed] [Google Scholar]
  17. Caffo O., Facchini G., Biasco E. Activity and safety of metronomic cyclophosphamide in the modern era of metastatic castration-resistant prostate cancer. Future Oncol. 2019;15(10):1115–1123. doi: 10.2217/fon-2018-0715. [DOI] [PubMed] [Google Scholar]
  18. Calcagno F., Mouillet G., Adotevi O. Metronomic cyclophosphamide therapy in hormone-naive patients withnon-metastatic biochemical recurrent prostate cancer: a phase II trial. Med. Oncol. 2016;33:8. doi: 10.1007/s12032-016-0806-0. [DOI] [PubMed] [Google Scholar]
  19. Calvan iN, Morelli F., Naglieri E. Metronomic chemotherapy with cyclophosphamide plus low dose of corticosteroids in advanced castration-resistant prostate cancer across the era of taxanes and new hormonal drugs. Med. Oncol. 2019;36(9):80. doi: 10.1007/s12032-019-1304-y. [DOI] [PubMed] [Google Scholar]
  20. Camerini A., Puccetti C., Donati S. Metronomic oral vinorelbine as first-line treatment in elderly patients with advanced non-small cell lung cancer: results of a phase II trial (MOVE trial) BMC Cancer. 2015;15:1–6. doi: 10.1186/s12885-015-1354-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Carreca, Bellomo I.U., Pernice F.M. Metronomic (M), capecitabine (C), and oxaliplatin (O) plus bevacizumab (B) as treatment of advanced colorectal cancer (ACRC) in very elderly people (M-COB): efficacy and safety (E&S) evaluation-A 2-year monitoring. ASCO Annual Meeting Proceedings. J. Clin. Oncol. 2011;29(15S):e14086. [Google Scholar]
  22. Cazzaniga M.E., Torri V., Villa F. Efficacy and safety of the all-oral schedule of metronomic vinorelbine and capecitabine in locally advanced or metastatic breast cancer patients: the Phase I-II VICTOR-1 Study. Int. J. Breast Cancer. 2014:769–790. doi: 10.1155/2014/769790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cazzaniga M.E., Cortesi L., Ferzi A. Metronomic chemotherapy with oral vinorelbine (mVNR) and capecitabine (mCAPE) in advanced HER2-negative breast cancer patients: is it a way to optimize disease control? Final results of the VICTOR-2 study. Breast Cancer Res. Treat. 2016;160(3):501–509. doi: 10.1007/s10549-016-4009-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chen C., Xu T., Lu Y. The efficacy of temozolomide for recurrent glioblastoma multiforme. Eur. J. Neurol. 2013;20:223–230. doi: 10.1111/j.1468-1331.2012.03778.x. [DOI] [PubMed] [Google Scholar]
  25. Chura J.C., Iseghem K., Van, Downs L.S. Bevacizumab plus cyclophosphamide in heavily pretreated patients with recurrent ovarian cancer. Gynecol. Oncol. 2007;107(2):326–330. doi: 10.1016/j.ygyno.2007.07.017. [DOI] [PubMed] [Google Scholar]
  26. Clarke J.L., Iwamoto F.M., Sul J. Randomized phase II trial of chemoradiotherapy followed by either dose-dense metronomic temozolomide for newly diagnosed glioblastoma. J. Clin. Oncol. 2009;27(23):3861–3867. doi: 10.1200/JCO.2008.20.7944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Colleoni M., Rocca A., Sandri M.T. Low-dose oral methotrexate and cyclophosphamide in metastatic breast cancer: antitumor activity and correlation with vascular endothelial growth factor levels. Ann. Oncol. 2002;13:73–80. doi: 10.1093/annonc/mdf013. [DOI] [PubMed] [Google Scholar]
  28. Colleoni M., Orlando L., Sanna G. A Metronomic low-dose oral cyclophosphamide and methotrexate plus or minus thalidomide in metastatic breast cancer: antitumor activity and biological effects. Ann. Oncol. 2006;17:232–238. doi: 10.1093/annonc/mdj066. [DOI] [PubMed] [Google Scholar]
  29. Colleoni M., Gray K.P., Gelber S. Low-dose oral cyclophosphamide and methotrexate maintenance for hormone receptor-negative early breast cancer: International Breast Cancer Study Group Trial 22-00. J. Clin. Oncol. 2016;34(28):3400–3408. doi: 10.1200/JCO.2015.65.6595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Correale P., Cerretani D., Remondo C. A novel metronomic chemotherapy regimen of weekly platinum and daily oral etoposide in high-risk non-small cell lung cancer patients. Oncol. Rep. 2006;16:133–140. doi: 10.3892/or.16.1.133. [DOI] [PubMed] [Google Scholar]
  31. Cremolini C., Marmorino F., Bergamo F. Phase II randomized study of maintenance treatment with bevacizumab or bevacizumab plus metronomic chemotherapy after first-line induction with FOLFOXIRI plus Bevacizumab for metastatic colorectal cancer patients: the MOMA trial. Eur. J. Cancer. 2019;109:175–182. doi: 10.1016/j.ejca.2018.12.028. [DOI] [PubMed] [Google Scholar]
  32. D’Ascanio M., Pezzuto A., Fiorentino C. Metronomic chemotherapy with vinorelbine produces clinical benefit and low toxicity in frail elderly patients affected by advanced non-small cell lung cancer. Biomed. Res. Int. 2018;27:1–6. doi: 10.1155/2018/6278403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Dabkara D., Ganguly S., Ghosh J. Metronomic cyclophosphamide in metastatic castrate resistant prostate cancer: experience from a tertiary cancer care center. Indian J. Cancer. 2018;55:94–97. doi: 10.4103/ijc.IJC_346_17. [DOI] [PubMed] [Google Scholar]
  34. De Iuliis F., Salerno G., Taglieri L. On and off metronomic oral vinorelbine in elderly women with advanced breast cancer. Tumori. 2015;101:30–35. doi: 10.5301/tj.5000207. [DOI] [PubMed] [Google Scholar]
  35. De la Haba J., Cazzaniga M., Freyer G. Randomised phase II study evaluating, as first-line chemotherapy, single-agent oral vinorelbine administered with two different schedules in patients with hormone receptor positive, HER2-negative advanced breast cancer (TempoBreast-1 trial). 38th Annual San Antonio Breast Cancer Symposium; San Antonio, TX, December 8–12; 2015. (abstr OT3-02-04) [Google Scholar]
  36. Dellapasqua S., Bertolini F., Bagnardi V. Metronomic cyclophosphamide and capecitabine combined with bevacizumab in advanced breast cancer. J. Clin. Oncol. 2008;26:4899–4905. doi: 10.1200/JCO.2008.17.4789. [DOI] [PubMed] [Google Scholar]
  37. Derosa L., Galli L., Orlandi P. Docetaxel plus oral metronomic cyclophosphamide: a phase II study with pharmacodynamic and pharmacogenetic analyses in castrationresistant prostate cancer patients. Cancer. 2014;120:3923–3931. doi: 10.1002/cncr.28953. [DOI] [PubMed] [Google Scholar]
  38. Di Desidero T., Derosa L., Galli L. Clinical, pharmacodynamic and pharmacokinetic results of a prospective phase II study on oral metronomic vinorelbine and dexamethasone in castration-resistant prostate cancer patients. Investig. New Drugs. 2016;34:760–770. doi: 10.1007/s10637-016-0385-0. [DOI] [PubMed] [Google Scholar]
  39. Eek D., Krohe M., Mazar I. Patient-reported preferences for oral versus intravenous administration for the treatment of cancer: a review of the literature. Patient Prefer Adher. 2016;10:1609–1621. doi: 10.2147/PPA.S106629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Ellebaek E., Engell-Noerregaard L., Iversen T.Z. Metastatic melanoma patients treated with dendritic cell vaccination. Interleukin-2 and metronomic cyclophosphamide: results from a phase II trial. Cancer Immunol. Immunother. 2012;61(10):1791–1804. doi: 10.1007/s00262-012-1242-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Fea E., Vanella P., Miraglio E. Metronomic oral cyclophosphamide (CTX) in patients (pts) with heavily pretreated metastatic castration-resistant prostate cancer (mCRPC), European Society for Medical Oncology Congress Proceedings. Ann. Oncol. 2016;2016(4S):iv38. [Google Scholar]
  42. Fedele P., Marino A., Orlando L. Efficacy and safety of low-dose metronomic chemotherapy with capecitabine in heavily pretreated patients with metastatic breast cancer. Eur. J. Cancer. 2012;48:24–29. doi: 10.1016/j.ejca.2011.06.040. [DOI] [PubMed] [Google Scholar]
  43. Ferrandina G., Corrado G., Mascilini F. Metronomic oral cyclophosphamide (MOC) in the salvage therapy of heavily treated recurrent ovarian cancer patients: a retrospective, multicenter study. BMC Cancer. 2014;14:1–7. doi: 10.1186/1471-2407-14-947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Fontana A., Galli L., Fioravanti A. Clinical and pharmacodynamic evaluation of metronomic cyclophosphamide, celecoxib, and dexamethasone in advanced hormone-refractory prostate cancer. Clin. Cancer Res. 2009;15:4954–4962. doi: 10.1158/1078-0432.CCR-08-3317. [DOI] [PubMed] [Google Scholar]
  45. Garcia A., AHirte H., Fleming G. Phase II clinical trial of bevacizumab and low-dosemetronomic oral cyclophosphamide in recurrent ovarian cancer: a trial of the California, Chicago, and Princess Margaret Hospital phase II consortia. J. Clin. Oncol. 2008;26(1):76–82. doi: 10.1200/JCO.2007.12.1939. [DOI] [PubMed] [Google Scholar]
  46. Garcia-Saenz J.A., Martin M., Calles A. Bevacizumab in combinationwith metronomic chemotherapy in patients with anthracycline- and taxane-refractory breast cancer. J. Chemother. 2008;20:632–639. doi: 10.1179/joc.2008.20.5.632. [DOI] [PubMed] [Google Scholar]
  47. Gebbia V., Serretta V., Borsellino N. Salvage therapy with oral metronomic cyclophosphamide andmethotrexate for castration-refractory metastatic adenocarcinoma of the prostate resistant to docetaxel. Urology. 2011;78:1125–1130. doi: 10.1016/j.urology.2011.08.010. [DOI] [PubMed] [Google Scholar]
  48. Glode L.M., Barqawi A., Crighton F. Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer. 2003;98:1643–1648. doi: 10.1002/cncr.11713. [DOI] [PubMed] [Google Scholar]
  49. Hanahan D., Bergers G., Bergsland E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target can target tumour angiogenesis in mice. J. Clin. Investig. 2000;105:1045–1047. doi: 10.1172/JCI9872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Handolias D., Quinn M., Foo S. Oral cyclophosphamide in recurrent ovarian cancer. Asia Pac. J. Clin. Oncol. 2016;12(1):e154–e160. doi: 10.1111/ajco.12074. [DOI] [PubMed] [Google Scholar]
  51. Hashimoto K., Man S., Xu Pet al. Potent preclinical impact of metronomic low-dose oral topotecan combined with the antiangiogenic drug pazopanib for the treatment of ovarian cancer. Mol Cancer Ther. 2010;9(4):996–1006. doi: 10.1158/1535-7163.MCT-09-0960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hatano K., Nonomura N., Nishimura K. Retrospective analysis of an oral combination of dexamethasone, uracil plus tegafur and cyclophosphamide for hormone-refractory prostate cancer. Jpn. J. Clin. Oncol. 2011;41:253–259. doi: 10.1093/jjco/hyq178. [DOI] [PubMed] [Google Scholar]
  53. Hellerstedt B., Pienta K.J., Redman B.G. Phase II trial of oral cyclophosphamide, prednisone, and diethylstilbestrol for androgen-independent prostate carcinoma. Cancer. 2003;98:1603–1610. doi: 10.1002/cncr.11686. [DOI] [PubMed] [Google Scholar]
  54. Jedeszko C., Paez-Ribes M., Di Desidero T. Postsurgical adjuvant or metastatic renal cell carcinoma therapy models reveal potent antitumor activity of metronomic oral topotecan with pazopanib. Sci. Transl. Med. 2015;7:282ra50. doi: 10.1126/scitranslmed.3010722. [DOI] [PubMed] [Google Scholar]
  55. Jellvert A., Lissbrant I.F., Edgren M. Effective oral combination metronomic chemotherapy with low toxicity for the management of castration-resistant prostate cancer. Exp. Therapeut. Med. 2011;2:579–584. doi: 10.3892/etm.2011.272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Jeong Y., Lee J.L. Efficacy of metronomic oral cyclophosphamide with low dose dexamethasone and celecoxib in metastatic castration-resistant prostate cancer. Asia-Pac. J. Clin. Oncol. 2017;13:204–211. doi: 10.1111/ajco.12583. [DOI] [PubMed] [Google Scholar]
  57. Jurado J.M., Sanchez A., Pajares B. Combined oral cyclophosphamide and bevacizumab in heavily pre-treatedovarian cancer. Clin. Transl. Oncol. 2008;10:583–586. doi: 10.1007/s12094-008-0254-7. [DOI] [PubMed] [Google Scholar]
  58. Kakolyris S., Samonis G., Koukourakis M. Treatment of non-small-cell lung cancer with prolonged oral etoposide. Am. J. Clin. Oncol. 1998;21:505–508. doi: 10.1097/00000421-199810000-00018. [DOI] [PubMed] [Google Scholar]
  59. Kamat A.A., Kim T.J., Landen C.N. Metronomic chemotherapy enhances the efficacy of antivascular therapy in ovarian cancer. Cancer Res. 2007;67:281–288. doi: 10.1158/0008-5472.CAN-06-3282. [DOI] [PubMed] [Google Scholar]
  60. Katsaounis P., Kotsakis A., Agelaki S. Cisplatin in combination with metronomic vinorelbine as front-line treatment in advanced non-small cell lung cancer: a multicenter phase II study of the hellenic oncology research group (HORG) Cancer Chemother. Pharmacol. 2015;75:821–827. doi: 10.1007/s00280-015-2707-x. [DOI] [PubMed] [Google Scholar]
  61. Kerbel R.S. Improving conventional or low dose metronomic chemotherapy with targeted antiangiogenic drugs. Cancer Res Treat. 2007;39(4):150–159. doi: 10.4143/crt.2007.39.4.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kesari S., Schiff D., Doherty L. Phase II study of metronomic chemotherapy for recurrent malignant gliomas in adults. Neuro-oncology. 2007;9(3):354–363. doi: 10.1215/15228517-2007-006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Khan O.A., Blann A.D., Payne M.J. Continuous low dose cyclophosphamide and methotrexate combined with celecoxib for patients with advanced cancer. Br. J. Cancer. 2011;104:1822–1827. doi: 10.1038/bjc.2011.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kong D.-S., Lee J.-I., Kim J.H. Phase II trial of low dose continuous (metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro-oncology. 2010;12(3):289–296. doi: 10.1093/neuonc/nop030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kontopodis E., Hatzidaki D., Varthalitis A phase II study of metronomic oral vinorelbine administered in the second line and beyond in non-small cell lung cancer (NSCLC): a phase II study of the hellenic oncology research group. J. Chemother. 2013;25:49–55. doi: 10.1179/1973947812Y.0000000050. [DOI] [PubMed] [Google Scholar]
  66. Kouroussis C., Vamvakas L., Vardakis N. Continuous administration of daily low-dose temozolomide in pretreated patients with advanced non-small cell lung cancer: a phase II study. Oncology. 2009;76(2):112–117. doi: 10.1159/000192586. [DOI] [PubMed] [Google Scholar]
  67. Kucukoner M., Isikdogan A., Yaman S. Oral etoposide for platinum-resistant and recurrent epithelial ovarian cancer: a study by the anatolian society of medical oncology. Asian Pac. J. Cancer Prev. 2012;13:3973–3976. doi: 10.7314/apjcp.2012.13.8.3973. [DOI] [PubMed] [Google Scholar]
  68. Ladoire S., Eymard J.C., Zanetta S. Metronomic oral cyclophosphamide prednisolone chemotherapy is an effective treatment for metastatic hormone-refractory prostate cancer after docetaxel failure. Anticancer Res. 2010;30:4317–4323. [PubMed] [Google Scholar]
  69. Lambertini M., Toss A., Passaro A. Cancer care during the spread of coronavirus disease 2019 (COVID-19) in Italy: young oncologists’ perspective. ESMO Open. 2020;5(2) doi: 10.1136/esmoopen-2020-000759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Langkjer S.T., Kenholm J., Jensen Dupont J. The NAME trial: a direct comparison of classical oral Navelbine versus Metronomic Navelbine in metastatic breast cancer. Future Oncol. 2019;15(22):2561–2570. doi: 10.2217/fon-2019-0124. [DOI] [PubMed] [Google Scholar]
  71. Liang W., Guan W., Chen R. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21(3):335–337. doi: 10.1016/S1470-2045(20)30096-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Lokich J. Capecitabine: fixed daily dose and continuous (noncyclic) dosing schedule. Cancer Investig. 2004;22:713–717. doi: 10.1081/cnv-200032976. [DOI] [PubMed] [Google Scholar]
  73. Lord R., Nair S., Schache A. Low dose metronomic oral cyclophosphamide for hormone resistant prostate cancer: a phase II study. J. Urol. 2007;177:2136–2140. doi: 10.1016/j.juro.2007.01.143. [DOI] [PubMed] [Google Scholar]
  74. Metronomic Chemotherapy of Capecitabine after Standard Adjuvant Chemotherapy in Operable Triple Negative Breast Cancer (MACRO). ClinicalTrials.gov Identifier: NCT02012634.
  75. Markman M., Hakes T., Reichman B. Phase 2 trial of chronic low-dose oral etoposide as salvage therapy of platinum refractory ovarian cancer. J. Cancer Res. Clin. Oncol. 1992;119:55–57. doi: 10.1007/BF01209489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Maulard-Durdux C., Dufour B., Hennequin C. Phase II study of the oral cyclophosphamide and oral etoposide combination in hormone refractory prostate carcinoma patients. Cancer. 1996;77:1144–1148. doi: 10.1002/(sici)1097-0142(19960315)77:6<1144::aid-cncr21>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  77. Mayer E.L., Isakoff S.J., Klement G. Combination antiangiogenic therapy in advanced breast cancer: a phase 1 trial of vandetanib, a VEGFR inhibitor, and metronomic chemotherapy, with correlative platelet proteomics. Breast Cancer Res. Treat. 2012;136:169–178. doi: 10.1007/s10549-012-2256-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Mayer E.L., Ligibel J.A., Burstein H.J. TBCRC 012L ABCDE, a phase II randomized study of adjuvant bevacizumab, metronomic chemotherapy (CM), diet and exercise after preoperative chemotherapy for breast cancer. ASCO Annual Meeting Proceedings. J. Clin. Oncol. 2016;28(15s):103. [Google Scholar]
  79. Mencoboni M., Filiberti R.A., Taveggia P. Safety of first-line chemotherapy with metronomic single-agent oral vinorelbine in elderly patients with NSCLC. Anticancer Res. 2017;37(6):3189–3194. doi: 10.21873/anticanres.11679. [DOI] [PubMed] [Google Scholar]
  80. Meng L.J., Wang J., Fan W.F. Evaluation of oral chemotherapy with capecitabine and cyclophosphamide plusthalidomide and prednisone in prostate cancer patients. J. Cancer Res. Clin. Oncol. 2012;138:333–339. doi: 10.1007/s00432-011-1101-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. MEtronomicTrEatment Option in Advanced bReastcAncer (METEORA-II). ClinicalTrials.gov Identifier: NCT02954055.
  82. Montagna E., Cancello G., Bagnardi V. Metronomic chemotherapy combined with bevacizumab and erlotinib in patients with metastatic HER2-negative breast cancer: clinical and biological activity. Clin. Breast Cancer. 2012;12:207–214. doi: 10.1016/j.clbc.2012.03.008. [DOI] [PubMed] [Google Scholar]
  83. Montagna E., BagnardiV, Cancello G. Metronomic chemotherapy for first-line treatment of metastatic triple-negative breast cancer: a phase II trial. Breast Care. 2018;13(3):177–181. doi: 10.1159/000487630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Muraki C., Ohga N., Hida Y. Cyclooxygenase-2 inhibition causes antiangiogenic effects on tumor endothelial and vascular progenitor cells. Int. J. Cancer. 2012;130:59–70. doi: 10.1002/ijc.25976. [DOI] [PubMed] [Google Scholar]
  85. Nasr K.E., Osman M.A., Elkady M.S. Metronomic methotrexate and cyclophosphamide after carboplatin included adjuvant chemotherapy in triple negative breast cancer: a phase III study. Ann. Transl. Med. 2015;3:284. doi: 10.3978/j.issn.2305-5839.2015.11.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Nelius T., Klatte T., De Riese W. Clinical outcome of patients with docetaxel-resistant hormone- refractory prostate cancer treated with second-line cyclophosphamide-based metronomic chemotherapy. Med. Oncol. 2010;27(2):363–367. doi: 10.1007/s12032-009-9218-8. [DOI] [PubMed] [Google Scholar]
  87. 2020. COVID-19 rapid guideline: delivery of systemic anticancer treatments NICE guideline. https://www.nice.org.uk/guidance/ng161 (Accessed 20 March 2020) [PubMed] [Google Scholar]
  88. Nishimura K., Nonomura N., Ono Y. Oral combination of cyclophosphamide, uracil plus tegafur and estramustine for hormone-refractory prostate cancer. Oncology. 2001;60:49–54. doi: 10.1159/000055296. [DOI] [PubMed] [Google Scholar]
  89. Noronha V., Joshi A., Marfatia S. Health-related quality of life in patients with metastatic, relapsed, or inoperable squamous cell carcinoma of the head and neck in India. Support. Care Cancer. 2016;24:1595–1602. doi: 10.1007/s00520-015-2937-9. [DOI] [PubMed] [Google Scholar]
  90. Omuro A., Chan A., Abrey L.E. Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant glioma. Neuro-oncology. 2013;15(2):242–250. doi: 10.1093/neuonc/nos295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Ontario Health, Cancer Care Ontario . 2020. Pandemic Planning Clinical Guideline for Patients with Cancer. https://www.accc-cancer.org/docs/document/cancer-program-fundamentals/oh-ccopandemic-planning-clinical-guidelines (Accessed 23 March 2020) [Google Scholar]
  92. Orlando L., Cardillo A., Ghisini R. Trastuzumab in combination with metronomic cyclophosphamide and methotrexate in patients with Her-2 positive metastatic breast cancer. BMC Cancer. 2006;6:225–230. doi: 10.1186/1471-2407-6-225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Orlando L. When less is better: safety and efficacy of combination of trastuzumab and continuous low oral dose chemotherapy (HEX) as first-line therapy for HER2-positive advanced breast cancer (ABC)—first early results from a phase II trial on behalf of Gruppo Oncologico Italia Meridionale (GOIM) J. Clin. Oncol. 2017;31(15s):618. [Google Scholar]
  94. Pai P.S., Vaidya A.D., Prabhash K. Oral metronomic scheduling of anticancer therapy-based treatment compared to existing standard of care in locally advanced oral squamous cell cancers: a matched-pair analysis. Indian J. Cancer. 2013;50(2):135–141. doi: 10.4103/0019-509X.117024. [DOI] [PubMed] [Google Scholar]
  95. Pandey A., Abhay D., Sunny J. Outcomes of advanced epithelial ovarian cancer with integration of metronomic chemotherapy: an Indian rural cancer centre experience. South Asian J. Cancer. 2016;5:59. doi: 10.4103/2278-330X.181627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Pandey A., Desai A., Ostwal V. Outcome of operable oral cavity cancer and impact of maintenance metronomic chemotherapy: a retrospective study from rural India. South Asian J. Cancer. 2016;5:52–55. doi: 10.4103/2278-330X.181625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Pasini F., Barile C., Caruso D. Oral Metronomic Vinorelbine (OMV) in elderly or pretreated patients with advanced non small cell lung cancer: outcome and pharmacokinetics in the real world. Investig. New Drugs. 2018;36(5):927–932. doi: 10.1007/s10637-018-0631-8. [DOI] [PubMed] [Google Scholar]
  98. Pastina P., Nardone V., Botta C. Radiotherapy prolongs the survival of advanced non-smallcell lung cancer patients undergone to an immune-modulating treatment with dosefractionedcisplatin and metronomic etoposide and bevacizumab (mPEBev) Oncotarget. 2017;8(44):75904–75913. doi: 10.18632/oncotarget.20411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Patil V.M., Noronha V., Joshi A. Metronomic chemotherapy in platinum-insensitive failures and/or early failures postmultimodality management in oral cancers. Indian J. Med. Paediatr. Oncol. 2015;36:161–165. doi: 10.4103/0971-5851.166725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Patil V.M., Noronha V., Joshi A. Phase I/II study of palliative triple metronomic chemotherapy in platinum refractory oral cancer. J. Clin. Oncol. 2019;37(32):3032–3041. doi: 10.1200/JCO.19.01076. [DOI] [PubMed] [Google Scholar]
  101. Peereboom D.M., Alban T.J., Grabowski M.M. Metronomic capecitabine as an immune modulator in glioblastoma patients reduces myeloid-derived suppressor cells. JCI Insight. 2019;4(22):1–15. doi: 10.1172/jci.insight.130748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Perry J.R., Belanger K., Mason W.P. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J. Clin. Oncol. 2010;28:2051–2057. doi: 10.1200/JCO.2009.26.5520. [DOI] [PubMed] [Google Scholar]
  103. Petrioli R., Roviello G., Laera L. Cisplatin, etoposide, and bevacizumab regimen followed by oral etoposide and bevacizumab maintenance treatment in patients with extensive stagesmall cell lung cancer: a single-institution experience. Clin. Lung Cancer. 2015;16(6):e229–e234. doi: 10.1016/j.cllc.2015.05.005. [DOI] [PubMed] [Google Scholar]
  104. Pfeiffer P., Rytte R.C., Madsen E.L. Five-day oral etoposide treatment for advanced small-cell lung cancer: Randomized comparison with intravenous chemotherapy. J. Natl. Cancer Inst. 1997;89:1892–1893. doi: 10.1093/jnci/89.24.1892. [DOI] [PubMed] [Google Scholar]
  105. Pujol J.L., Coffy A., Camerini A. An individual patient data meta-analysis of metronomic oral Vinorelbine in metastatic non small cell lung cancer. PLOS ONE. 2019;14(8):e0220988. doi: 10.1371/journal.pone.0220988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Reardon D.A., Desjardins A., Vredenburgh J.J. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a Phase II Study. Br. J. Cancer. 2009;101(12):1986–1994. doi: 10.1038/sj.bjc.6605412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Reardon D.A., Desjardins A., Peters K. Phase II study of metronomic chemotherapy with bevacizumab for recurrent glioblastoma after progression on bevacizumab therapy. J. Neurooncol. 2011;103:371–379. doi: 10.1007/s11060-010-0403-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Revannasiddaiah S., Joshi S.C., Pandey K.C. The results with the addition of metronomic cyclophosphamide to palliative radiotherapy for the treatment of non-small cell lung carcinoma. Ann. Transl. Med. 2015;3(20):1–7. doi: 10.3978/j.issn.2305-5839.2015.11.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Robles C., Furst A.J., Sriratana P. Phase II study of vinorelbine with low dose prednisone in the treatment of hormone-refractory metastatic prostate cancer. Oncol. Rep. 2003;10:885–889. doi: 10.3892/or.10.4.885. [DOI] [PubMed] [Google Scholar]
  110. Rochlitz C., Bigler M., von Moos R. SAKK 24/09: safety and tolerability of bevacizumab plus paclitaxel vs. bevacizumab plus metronomic cyclophosphamide and capecitabine as first-line therapy in patients with HER2-negative advanced stage breast cancer – a multicenter, randomized phase III trial. BMC Cancer. 2016;16(1):780. doi: 10.1186/s12885-016-2823-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Romiti A., Onesti C.E., Roberto M. Continuous, low-dose capecitabine for patients with recurrent colorectal cancer. Med. Oncol. 2015;32:54. doi: 10.1007/s12032-015-0496-z. [DOI] [PubMed] [Google Scholar]
  112. Romiti A., Falcone R., Roberto M., Marchetti P. Current achievements and future perspectives of metronomic chemotherapy. Investig. New Drugs. 2017;35(3):359–374. doi: 10.1007/s10637-016-0408-x. [DOI] [PubMed] [Google Scholar]
  113. Salem D.A., Gado N.M., Abdelaziz N.N. Phase II trial of metronomic chemotherapy as salvage therapy for patients with metastatic breast cancer. J. Egypt Natl. Cancer Inst. 2008;20:134–140. [PubMed] [Google Scholar]
  114. Samaritani R., Corrado G., Vizza E. Cyclophosphamide “metronomic” chemotherapy for palliative treatment of a young patient with advanced epithelial ovarian cancer. BMC Cancer. 2007;7:54. doi: 10.1186/1471-2407-7-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Sanchez-Munoz A., Mendiola C., Perez-Ruiz E. Bevacizumab plus low-dose metronomic oral cyclophosphamide in heavily pretreated patients with recurrent ovarian cancer. Oncology. 2010;79:98–104. doi: 10.1159/000320602. [DOI] [PubMed] [Google Scholar]
  116. Schwartzberg L.S., Wang G., Somer B.G. Phase II trial of fulvestrant with metronomic capecitabine for postmenopausal women with hormone receptor-positive, HER2-negative metastatic breast cancer. Clin. Breast Cancer. 2014;14:13–19. doi: 10.1016/j.clbc.2013.09.003. [DOI] [PubMed] [Google Scholar]
  117. Sharma A., Malik P.S., Kumar L. Oral metronomic chemotherapy for recurrent and refractory epithelial ovarian cancer: a retrospective analysis. Indian J. Med. Res. 2019;150(6):575–583. doi: 10.4103/ijmr.IJMR_2030_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Shawky H., Galal S. Preliminary results of capecitabine metronomic chemotherapy in operable triple-negative breast cancer after standard adjuvant therapy – a single-arm phase II study. J. Egypt Natl. Cancer Inst. 2014;26:195–202. doi: 10.1016/j.jnci.2014.10.002. [DOI] [PubMed] [Google Scholar]
  119. Shi H., Jiang J., Ji J., Shi M., Cai Q., Chen X. Anti-angiogenesis participates in antitumor effects of metronomic capecitabine on colon cancer. Cancer Lett. 2014;349:128–135. doi: 10.1016/j.canlet.2014.04.002. [DOI] [PubMed] [Google Scholar]
  120. Shi M., Ma T., Xi W. A study of capecitabine metronomic chemotherapy is non-inferior to conventional chemotherapy as maintenance strategy in responders after induction therapy in metastatic colorectal cancer. Trials. 2020;21:249–255. doi: 10.1186/s13063-020-4194-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Shitara K., Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann. N. Y. Acad. Sci. 2018;1417:104–115. doi: 10.1111/nyas.13625. [DOI] [PubMed] [Google Scholar]
  122. Simeone E., Daponte A., De Feo G. “Metronomic schedule of temozolomide with conventional dose of cisplatin in metastatic melanoma,” ASCO Annual Meeting Proceedings. J. Clin. Oncol. 2009;27(15S) [Google Scholar]
  123. Simkens L.H.J., Van Tinteren H., May A. Maintenance treatment with capecitabine and bevacizumab in metastatic colorectal cancer (CAIRO3): a phase 3 randomised controlled trial of the dutch colorectal cancer group. Lancet. 2015;385:1843–1852. doi: 10.1016/S0140-6736(14)62004-3. [DOI] [PubMed] [Google Scholar]
  124. Stockhammer F., Misch M., Koch A. Continuous low dose temozolomide and celecoxib in recurrent glioblastoma. J. Neurooncol. 2010;100:407–415. doi: 10.1007/s11060-010-0192-y. [DOI] [PubMed] [Google Scholar]
  125. Stockler M.R., Harvey V.J., Francis P.A. Capecitabine versus classical cyclophosphamide, methotrexate, and fluorouracil as first-line chemotherapy for advanced breast cancer. J. Clin. Oncol. 2011;29:4498–4504. doi: 10.1200/JCO.2010.33.9101. [DOI] [PubMed] [Google Scholar]
  126. Surmont V., Aerts J.G., Pouw E. Oral UFT, etoposide and leucovorin in recurrent non-small cell lung cancer: a nonrandomized phase II study. Lung Cancer. 2009;66:333–337. doi: 10.1016/j.lungcan.2009.02.016. [DOI] [PubMed] [Google Scholar]
  127. Tan E.H., Tan D.S.W., Li W.Y. Metronomic vinorelbine (oral) in combination with sorafenib in advanced non-small cell lung cancer. Lung Cancer. 2015;88:289–296. doi: 10.1016/j.lungcan.2015.04.001. [DOI] [PubMed] [Google Scholar]
  128. Tanaka H., Matsushima H., Nishibu A., Clausen B.E., Takashima A. Dual therapeutic efficacy of vinblastine as a unique chemotherapeutic agent capable of inducing dendritic cell maturation. Cancer Res. 2009;69:6987–6994. doi: 10.1158/0008-5472.CAN-09-1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Tralongo S., Bordonaro A., Di Mari Chemotherapy in frail elderly patients with hormone-refractory prostate cancer: a “real world” experience. Prostate Int. 2016;4(1):15–19. doi: 10.1016/j.prnil.2015.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Tupikowski K., Dembowski J., Kolodziej A. Interferon alpha and metronomic cyclophosphamide for metastatic kidney cancer: a phase 2 study. J. Interferon Cytokine Res. 2015;35(5):367–372. doi: 10.1089/jir.2014.0106. [DOI] [PubMed] [Google Scholar]
  131. Ugurel S., Rappl G., Tilgen W. Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J. Clin. Oncol. 2001;19:577–583. doi: 10.1200/JCO.2001.19.2.577. [DOI] [PubMed] [Google Scholar]
  132. Van Cutsem E., Findlay M., Osterwalder B. Capecitabine, an oral fluoropyrimidine carbamate with substantial activity in advanced colorectal cancer: results of a randomized phase II study. J. Clin. Oncol. 2000;18:1337–1345. doi: 10.1200/JCO.2000.18.6.1337. [DOI] [PubMed] [Google Scholar]
  133. Van Dodewaard-de Jong J.M., Verheul H.M., Bloemendal H.J. New treatment options for patients with metastatic prostate cancer: what is the optimal sequence? Clin. Genitourinary Cancer. 2015;13:271–279. doi: 10.1016/j.clgc.2015.01.008. [DOI] [PubMed] [Google Scholar]
  134. Vincent I., Mignot G., Chalmin 5FU selectively kills tumor associated myeloid derived suppressor cells resulting in enhanced T cell dependent antitumor immunity. Cancer Res. 2010;69:3052–3061. doi: 10.1158/0008-5472.CAN-09-3690. [DOI] [PubMed] [Google Scholar]
  135. Walter B., Schrettenbrunner I., Vogelhuber M. Pioglitazone, etoricoxib, interferon-a, andmetronomiccapecitabine for metastatic renal cell carcinoma: final results of a prospective phase II trial. Med. Oncol. 2012;29(2):799–805. doi: 10.1007/s12032-011-9982-0. [DOI] [PubMed] [Google Scholar]
  136. Wang Z., Lu J., Leaw S. An all-oral combination of metronomic cyclophosphamide plus capecitabine in patients with anthracycline and taxane-pretreated metastatic breast cancer: a phase II study. Cancer Chemother. Pharmacol. 2012;69:515–522. doi: 10.1007/s00280-011-1728-3. [DOI] [PubMed] [Google Scholar]
  137. Wang Z., Wang J., He J. Active and effective measures for the care of patients with cancer during the COVID-19 spread in China. JAMA Oncol. 2020 doi: 10.1001/jamaoncol.2020.1198. [DOI] [PubMed] [Google Scholar]
  138. Watanabe T., Sano M., Takashima S. Oral uracil and tegafur compared with classic cyclophosphamide, methotrexate, fluorouracil as postoperative chemotherapy in patients with node-negative, high-risk breast cancer: National Surgical Adjuvant Study for Breast Cancer 01 Trial. J. Clin. Oncol. 2009;27(9):1368–1374. doi: 10.1200/JCO.2008.18.3939. [DOI] [PubMed] [Google Scholar]
  139. Welzel G., Giordano F.A., Gehweiler J. Metronomic chemotherapy with daily low-dose temozolomide and celecoxib in elderly patients with newly diagnosed glioblastoma multiforme: a retrospective analysis. J. Neurooncol. 2015;124(2):265–273. doi: 10.1007/s11060-015-1834-x. [DOI] [PubMed] [Google Scholar]
  140. Wong C., Liu F.S. Continuous oral cyclophosphamide as salvage or maineinance therapy in ovarian, primary peritoneal, and fallopian tube cancers: a retrospective single institute experience. Taiwan. J. Obstret. Gynecol. 2017;56:302–305. doi: 10.1016/j.tjog.2017.04.006. [DOI] [PubMed] [Google Scholar]
  141. Yashi M., Nishihara D., Mizuno T. Metronomic oral cyclophosphamide chemotherapy possibly contributes to stabilization of disease in patients with metastatic castration resistant prostate cancer: a prospective analysis of consecutive cases. Clin. Genitourinary Cancer. 2014;12(5):e197–e203. doi: 10.1016/j.clgc.2014.02.007. [DOI] [PubMed] [Google Scholar]
  142. Yoshimoto M., Takao S., Hirata M. Metronomic oral combination chemotherapy with capecitabine and cyclophosphamide: a phase II study in patients with HER2-negative metastatic breast cancer. Cancer Chemother. Pharmacol. 2012;70:331–338. doi: 10.1007/s00280-012-1826-x. [DOI] [PubMed] [Google Scholar]
  143. You B., Ravaud A., Canivet A. The official French guidelines to protect patients with cancer against SARS-CoV-2 infection. Lancet Oncol. 2020 doi: 10.1016/S1470-2045(20)30204-7. S1470-2045(20):30204-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Zustovich F., Landi L., Lombardi G. Sorafenib plus daily low-dose temozolomide for relapsed glioblastoma: a phase II study. Anticancer Res. 2013;33(8):3487–3494. [PubMed] [Google Scholar]

Articles from Critical Reviews in Oncology/Hematology are provided here courtesy of Elsevier

RESOURCES