2,362
Views
4
CrossRef citations to date
0
Altmetric
Review

Prophylaxis and treatment strategies for optimizing chemotherapy relative dose intensity

, & ORCID Icon
Pages 1145-1159 | Received 07 Apr 2021, Accepted 09 Jun 2021, Published online: 21 Jun 2021

ABSTRACT

Introduction

A decrease in relative-dose intensity (RDI) of chemotherapy has been shown to be associated with poor patient outcomes in solid tumors and non-Hodgkin’s lymphoma. The actual delivered chemotherapy dose received by patients can be influenced by dose reductions and treatment delays, often due to toxicities, most commonly chemotherapy-induced neutropenia (CIN).

Areas covered

We review seminal evidence and more recent studies that have shown an association between higher RDI and improved patient survival. A smaller number of studies has shown no association between RDI and outcomes. These differences may be due to study limitations, including low power, differences in patient and disease characteristics, or the chemotherapeutic regimen. We describe guidelines recommendations to prevent and treat CIN with granulocyte-colony stimulating factor (G-CSF) and describe novel approaches to prevent neutropenia that are being developed that may provide greater value and be associated with fewer adverse events than standard G-CSF options.

Expert opinion

Maintaining RDI is important to ensure optimal patient outcomes. This can be achieved through the proper administration of G-CSF prophylaxis and treatment. Newer agents in development to treat and/or prevent CIN are entering regulatory review and may potentially change the treatment landscape for CIN in the future.

1. Introduction

The relative dose intensity (RDI) of chemotherapy has been shown to impact outcomes of patients with early stage solid tumors and lymphoma, with low RDI associated with shorter survival. RDI is defined as the ratio of the delivered dose intensity to the reference dose intensity for the specific chemotherapy regimen [Citation1–3]. The reported RDI threshold to achieve optimal patient outcomes varies by cancer type and is suggested to be 85% for early stage breast cancer and ~70% for non-Hodgkin’s lymphoma (NHL) [Citation4,Citation5]. Deviations in optimal RDI are often due to dose reductions, treatment delays, or withdrawal from therapy due to adverse events experienced throughout chemotherapy [Citation2,Citation6,Citation7]. Chemotherapy-induced neutropenia (CIN) is the most common adverse event leading to reductions in RDI, thereby negatively impacting clinical outcomes. Accordingly, healthcare costs are also increased due to hospitalizations for patients with febrile neutropenia and related complications [Citation7–11].

The incidence of CIN or febrile neutropenia varies across tumor types and chemotherapy regimens [Citation8,Citation9,Citation12–15], with rates of 0‒48% reported for breast cancer, 3‒57% for small cell lung cancer, 0‒54% for non-small cell lung cancer (NSCLC), and 10‒78% for NHL [Citation12]. Across all tumor types, neutropenia is more common during the first cycle of treatment when patients typically receive the full dose(s) of chemotherapy and decreases with subsequent cycles of treatment, irrespective of disease stage, patient age, and line of therapy [Citation8]. The incidence of neutropenia tends to decrease from cycle 2 onward as physicians try to manage it with preventative strategies [Citation8,Citation14].

Severe neutropenia (grade 3 or 4) is a significant complication that can lead to increased morbidity and mortality, which also varies according to tumor type [Citation2,Citation9,Citation13–15]. In a retrospective analysis of 5,990 patients with solid tumors or NHL compared to propensity score–matched controls, the incidence of early and overall mortality was higher in patients who developed febrile neutropenia compared to those who did not (HR 1.15 and 1.35, respectively). Patients with lung cancer had the greatest risk of early mortality, while the risk was lowest for patients with breast cancer [Citation13]. In general, mortality rates for patients hospitalized for febrile neutropenia are approximately 10% and even higher for patients with multiple or severe comorbidities (approximately 20%) [Citation2,Citation14,Citation16].

Patients who develop CIN may also require hospitalization [Citation9,Citation13,Citation14], with mean length of hospital stay ranging from 4.1 to 7.9 days [Citation9]. This increase in hospitalization and patient management has a large economic impact [Citation9,Citation15]. A retrospective analysis of patients treated between 2006 and 2009 reported total costs for NHL, lung or breast cancer patients who experienced febrile neutropenia as 37,555, USD 32,964, USD and 20,462, USD respectively (in 2013 USD) [Citation9]. Similarly, according to an analysis of the 2012 National Inpatient Sample and Kids’ Inpatient Database, the total cost for 108,419 patients hospitalized for cancer-related neutropenia in 2012 was 2.7 USD billion (in 2012 USD) [Citation17].

To manage CIN, physicians often dose-reduce or delay chemotherapy to allow for neutrophil recovery. Although this approach reduces the negative impact of chemotherapy, it also results in lower RDI and poorer clinical outcomes because patients are not receiving the optimal dose and frequency of chemotherapy to most effectively treat their disease [Citation2,Citation6,Citation7]. Administration of granulocyte-colony stimulating factor (G-CSF) therapy is commonly used and recommended by guidelines for patients who have a high risk of neutropenia to prevent the development of CIN, which allows maintenance of optimal RDI and improved patient outcomes [Citation12,Citation18–23]. However, because the currently available G-CSF therapies are associated with adverse events, such as bone pain, headache, and myalgias, there is interest in treatments that are effective in preventing CIN while reducing adverse events and health care costs [Citation22,Citation24]. Furthermore, some patients receiving G-CSF will still experience febrile neutropenia; therefore, there is a need for continued investigations of drugs other than G-CSF to further reduce the risk of febrile neutropenia [Citation25].

2. CIN and its impact on relative dose intensity

Use of chemotherapy regimens with myelosuppressive properties, such as cyclophosphamide/methotrexate/fluorouracil (CMF), fluorouracil/doxorubicin/ cyclophosphamide, or docetaxel/cyclophosphamide (TC), in early stage breast cancer can increase the risk of CIN and is associated with an increased risk of low RDI [Citation6]. Among 20,799 early stage breast cancer patients treated with adjuvant chemotherapy from 1997‒2000, dose reductions >15% and treatment delays >7 days occurred in 37% and 25% of patients, respectively. This resulted in 56% of patients receiving an RDI <85%. Interestingly, despite the high rates of dose delays and reductions, G-CSF was only administered in 26% of patients. Furthermore, G-CSF use was predominantly reactive or therapeutic – and not prophylactic – as rates of administration were highest in cycles 3 and 4 [Citation6].

This association between the occurrence of CIN and increased dose delays/reductions and subsequent low RDI has also been observed in other studies in early stage breast cancer, solid tumors, and NHL [Citation7,Citation11,Citation26]. Recent studies have revealed that CIN, dose delays/reductions, and low RDI are still common among early stage breast cancer and NHL patients within the past decade; however, they tend to be lower than what was reported in the early 2000s [Citation8,Citation10,Citation27–30]. This has been attributed to higher rates of G-CSF administration in the past two decades. For example, a comparison of aggressive B-cell NHL patients treated between 2006‒2009 and 1993‒2001 found fewer patients experienced dose reductions (21% vs 35%, respectively) and the percent of patients achieving RDI ≥85% was higher (68% vs 52%, respectively) in more recent years. This was associated with a lower incidence of febrile neutropenia (12% vs 21%) and febrile neutropenia–related hospitalization (10% vs 16%), and higher rates of G-CSF use (75% vs 12%) during the period from 2006–2009 [Citation30]. These combined observations suggest that oncologists may be more aware of recommended strategies to prevent and treat CIN than they were in the past [Citation10,Citation30].

While low RDI due to CIN is common across solid tumor types and lymphomas, its risk varies among patients. The most important risk factor for the development of CIN is the type and intensity of myelosuppressive chemotherapy used. Additional factors found to be associated with increased risk of CIN include age >65 years, poor performance status, comorbidities, advanced disease, history of prior febrile neutropenia, and no prophylactic antibiotic or G-CSF use [Citation2,Citation12]. Of note, these factors are also reported to be associated with low RDI in breast cancer and NHL patients [Citation6,Citation7,Citation26].

3. The association between RDI and outcomes in solid tumors and lymphomas

3.1. The importance of dose intensity

Evidence demonstrates that dose intensity of adjuvant chemotherapy is an important predictor of clinical outcomes. This has primarily been noted in studies of early stage breast cancer and other solid tumors [Citation31,Citation32], with high or moderate dose intensity chemotherapy associated with longer survival [Citation33]. A recent meta-analysis of 26 trials of 37,298 patients with breast cancer found that increasing the dose density of adjuvant chemotherapy by shortening the interval between treatment cycles reduces the 10-year risk of recurrence and death, without increasing mortality from other causes [Citation32]. Improved outcomes have also been observed in bladder cancer patients receiving higher dose intensity chemotherapy [Citation34–36]. It is important to note that these dose-dense treatment strategies cannot be effectively delivered without the use of G-CSF [Citation31].

Several hypotheses have been proposed to explain the benefit of higher dose intensity regimens. Among these, the Norton–Simon hypothesis suggests that more frequent administration of chemotherapy can minimize residual tumor burden. According to Gompertzian kinetics models, chemotherapy that reduces tumor volume will result in an inherent increase in tumor regrowth between cycles. Therefore, to enhance cytoreductive activity, chemotherapy should be delivered in the shortest interval possible. Furthermore, the Goldie–Coldman hypothesis suggests administration of high intensity dose regimens prevents the accumulation of mutations that could lead to drug-resistant clones and subsequently reduce chemotherapy effect [Citation31].

3.2. High RDI is associated with improved outcomes

In addition to dose intensity, high RDI has been shown to be associated with improved patient outcomes across many tumor types. This association has been extensively studied in early stage breast cancer and NHL, with similar observations found in other tumor types [Citation2]. This relationship between high RDI and improved outcomes is particularly strong in patients with early stage disease treated with regimens delivered with curative intent [Citation2]; however, this association is less clear in advanced/metastatic disease [Citation37], which is beyond the scope of this review. Furthermore, there is limited evidence that dosing above the recommended RDI (>100%) improves outcomes, likely due to added toxicities with higher doses [Citation38,Citation39].

The importance of RDI in regard to patient outcomes was shown in a seminal study that enrolled early stage breast cancer patients from 1973‒1975 treated with CMF. After a median follow-up of 20 and 30 years, patients who received a higher RDI (≥85%) had improved relapse-free survival (RFS) and overall survival (OS) [Citation4,Citation40]. More recent prospective and retrospective analyses in early stage breast cancer have confirmed these historical findings, with higher RDI associated with longer OS and a lower risk of death [Citation41–43]. Higher RDI has also been reported to be associated with improved survival in other solid tumors, including colon [Citation44,Citation45], ovarian [Citation38,Citation39], and pancreatic ductal adenocarcinoma [Citation46]. Data from these studies are summarized in .

Table 1. Summary of studies of RDI and patient outcomes in solid tumors and NHL

Similar associations between RDI and outcomes have also been reported in NHL (). Early studies in NHL patients treated with an anthracycline-containing induction regimen showed that 28% of patients had an RDI <70%, which was associated with a decreased response rate and shorter overall 2-year survival [Citation5]. Similar findings have been reported in more recent studies with the rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) regimen, noting poor survival associated with RDI of less than 50‒90% () [Citation47–50].

3.3. Optimal RDI cutoff values

In general, an RDI cutoff of 85% has been suggested to be optimal for early stage breast cancer patients [Citation4]; however, optimal RDI values may differ according to subtype. An analysis of 494 ER+/PR+, HER2‒ and 180 triple-negative breast cancer (TNBC) patients treated with standard chemotherapy in 2011 found that RDI <85% was associated with poor outcomes in ER+/PR+ HER2‒ patients and <75% in TNBC patients [Citation43]. This study was the first to show differences in RDI cutoff values among breast cancer subtypes, and it is unclear as to the reason for these differences; therefore, additional studies are needed to confirm these findings.

Differences in optimal RDI cutoff values have also been noted among NHL studies. Early observations suggested that RDI <70% was associated with poor outcomes [Citation5]; however, among recent studies assessing RDI in patients treated with R-CHOP21, optimal RDI values were reported as 70%, 85%, and 90% [Citation47,Citation49–51]. These variations may be due to differences in patient- or disease-specific characteristics among the cohorts, length of follow-up, or RDI calculation methods.

3.4. Studies showing no association between RDI and outcomes

Although many studies have shown improved survival with high RDI, some have reported no association between RDI and patient outcomes () [Citation52–55]. Explanations for these differing conclusions may include length of follow-up period, sample size, RDI calculation method, or disease- or patient-specific characteristics. Alternatively, the type of chemotherapy regimen could differentially impact potential associations between RDI and outcomes. The myelosuppressive effect of chemotherapy agents can differ, even among the same class of agents, as exemplified by taxanes. The incidence of neutropenia following docetaxel given every 3 weeks is much higher than weekly or every-3-weekly infusions of paclitaxel (46% vs ≤4%) [Citation56]. Thus, higher rates of dose delays/reductions and lower RDI levels are observed in docetaxel-treated patients [Citation41]. Furthermore, the impact of RDI may vary according to individual agents within a regimen or the duration of treatment [Citation41,Citation45,Citation51,Citation52]. Among HER2-negative breast cancer patients treated with docetaxel/doxorubicin/cyclophosphamide (TAC), an RDI ≥90% was associated with improved OS; however, an RDI ≥90% for the docetaxel-only portion of the regimen had no impact on OS, while RDI ≥90% for the doxorubicin/cyclophosphamide (AC) portion moderately impacted survival [Citation41]. Similarly, in NHL, reductions in RDI >15% were significantly associated with poor OS and progression-free survival (PFS) in patients treated with R-CHOP21; however, no associations were found between reductions in RDI and outcomes with the R-CHOP14 regimen [Citation51] ().

3.5. RDI in special populations

Elderly patients have an increased risk of toxicity and mortality during treatment with curative chemotherapy, and advanced age is considered the most important patient-specific risk factor for the development of febrile neutropenia [Citation2,Citation22]. As a result, elderly patients tend to receive more dose reductions and delays than do younger patients. Delays due to treatment-related toxicities result in lower RDI [Citation2]. Among 976 patients with solid tumors or lymphoma in a prospective analysis, 51% of patients age 70‒74 years received RDI ≤85%; this rate increased to 60% for patients age ≥80 years due to higher rates of dose delays and reductions. In the 50% of patients who received RDI ≥85%, there was no difference in the rates of severe or febrile neutropenia by age, suggesting advanced age alone does not increase the risk of hematologic toxicity [Citation57]. Similar observations have been reported in other studies; nevertheless, advanced age (≥65 years) remains a common variable associated with reduced chemotherapy RDI [Citation6,Citation7,Citation26,Citation53,Citation58].

Despite the risk of chemotherapy-induced toxicities, elderly patients benefit from chemotherapy to the same extent as younger patients; consequently, current guidelines do not recommend dose modification in the first cycle simply based on patient age [Citation2]. Several studies have confirmed that elderly patients are able to maintain RDI ≥85%, and appropriate patient assessment and prophylactic use of G-CSF agents could support optimal dosing [Citation53,Citation58]. A recent retrospective analysis of Medicare data of patients 66 years of age and older with breast cancer, lung cancer, or NHL who received myelosuppressive chemotherapy between 1995 and 2015 reported the use of prophylactic G-CSF increased after 2002 and was associated with an annual decrease in the incidence of neutropenia-related hospitalizations up to 2010, which then remained stable until 2015. These observations demonstrate that G-CSF administration is feasible and provides benefits in an elderly population [Citation59]. These benefits of G-CSF in elderly patients are reflected in guideline recommendations. Current National Comprehensive Cancer Network (NCCN) Guidelines consider growth factor support the best strategy to improve treatment in elderly patients and recommend G-CSF administration in this population when dose intensity is required for response or cure [Citation60].

In addition to advanced age, body surface area ≥2 m2 or high body mass index (BMI) is also associated with lower RDI [Citation6,Citation26,Citation61]. In a retrospective analysis of breast cancer patients, most of the difference in dose intensity in obese patients was in the intended dose, with no reductions due to toxicity. This suggests physicians routinely underdose overweight or obese patients [Citation6]. It has been reported that up to 40% of obese patients receive limited chemotherapy based on reduced intended dose [Citation62]. These reductions are initiated due to concerns regarding toxicities in this population; however, these concerns are unfounded, as numerous studies have shown no association between increased BMI and toxicities [Citation63–67]. Moreover, some studies have reported that the incidence of febrile neutropenia, reduced leukocyte count, and grade 3/4 toxicities are lower in obese patients receiving full-dose chemotherapy [Citation63,Citation64,Citation67]. Given these observations, American Society of Clinical Oncology (ASCO) guidelines recommend full weight-based chemotherapy dosing in obese patients, particularly in the curative setting, to maintain RDI and chemotherapy efficacy [Citation62].

4. Current strategies to prevent and Treat CIN, and improve RDI

Identification of patients at increased risk of developing CIN and appropriate prophylactic use of G-CSF to decrease risk of CIN is crucial to achieve optimal RDI levels, maintain chemotherapeutic activity, and prevent CIN-associated morbidity and mortality [Citation2,Citation12,Citation22,Citation23]. According to expert guidelines, maintenance of dose intensity with G-CSF should be standard of care for any treatment with a curative intent [Citation68]. G-CSF agents have been the standard of care in the prevention and management of CIN since the approval of the recombinant human G-CSF (rhG-CSF) drug filgrastim in 1991 [Citation69]. CIN supportive care improved in 2002 with the introduction of the first long-acting recombinant G-CSF, pegylated filgrastim, which allowed once-per-cycle therapy [Citation70]. Current treatment options for CIN have not changed substantially since that time, with common agents limited to rhG-CSF (filgrastim, tbo-filgrastim), long-acting pegylated rhG-CSF (pegfilgrastim), and their biosimilars (filgrastim-sndz, filgrastim-aafi, pegfilgrastim-jmdb, pegfilgrastim-dbqv, pegfilgrastim-bmez, pegfilgrastim-apgf) [Citation12,Citation22,Citation23]. While biosimilar products are highly similar to their reference product, they are not currently considered interchangeable and cannot be substituted without intervention of the prescriber [Citation71]. Furthermore, it is important to note that tbo-filgrastim was approved in Europe as a biosimilar in 2008; however, in the United States, it was approved in 2012 as a biologic product because it was filed under a Biologics License Application since the biosimilars pathways was not established at the time of FDA submission [Citation72].

Short-acting rhG-CSF agents (ie, filgrastim, tbo-filgrastim) are recommended to be administered daily 24 hours after cytotoxic chemotherapy for up to 2 weeks or until neutrophil recovery [Citation73,Citation74]. Alternatively, long-acting pegylated rhG-CSF agents (ie, pegfilgrastim) are administered once per cycle 24 hours post-chemotherapy [Citation70]. Both formulations can also be administered up to 3‒4 days after completion of myelosuppressive chemotherapy [Citation22]. Delayed administration of G-CSF ≥24 hours post-chemotherapy is recommended because G-CSF agents can induce the proliferation of myeloid progenitor cells. These cells can be negatively impacted by cytotoxic therapies, thereby further increasing the risk of febrile neutropenia. In fact, patients who receive G-CSF off schedule have an increased risk of developing febrile neutropenia [Citation75,Citation76].

Numerous studies, including meta-analyses, have shown that G-CSF agents reduce the incidence and duration of severe neutropenia and increase chemotherapy completion and RDI; however, effects on overall mortality and infection-related mortality are not as clear [Citation12]. A recent exploratory analysis of the phase 3 PANTHER study of 5-fluorouracil/epirubicin/cyclophosphamide plus docetaxel (FEC/D) versus EC/D adjuvant chemotherapy in breast cancer patients demonstrated the value of G-CSF therapy. Patients administered G-CSF had a lower incidence of neutropenic events and dose delays [Citation77]. The benefit of G-CSF agents has also been shown in meta-analyses of randomized, controlled trials. In all analyses, G-CSF agents reduced the risk of febrile neutropenia [Citation18,Citation19,Citation21,Citation78]. G-CSF was also shown to reduce hospitalization time and improve neutrophil recovery [Citation18,Citation21]; however, its impact on infection-related mortality and overall mortality varied among studies [Citation18–21].

Despite the benefits of G-CSF agents, they are also associated with several limitations, including potential adverse events and additional unplanned costs. Bone pain is the primary adverse event associated with G-CSF use, which has been reported in 25‒83% of patients across different tumor types and chemotherapeutic regimens. Additional common adverse events of G-CSF agents and their respective incidence rates include headache (15‒83%), myalgias (13‒68%), fatigue (9‒59%), nausea and/or vomiting (3‒18%), fever/chills/sweats (0‒27%), and skin reaction (1‒3%) [Citation22,Citation24]. Furthermore, rare serious adverse events may also occur during G-CSF administration, and all G-CSF agents include a warnings and precautions statement for these events, including fatal splenic rupture, acute respiratory distress syndrome, serious allergic reactions, sickle cell crisis in patients with sickle cell disease, glomerulonephritis, and capillary leak syndrome [Citation24,Citation73,Citation74,Citation79]. While these events are rare, physicians need to be aware of them and act accordingly to reduce the risk of their occurrence. In addition to these potential adverse events, the need for regular clinical visits to receive G-CSF may be challenging for some patients; however, on-body injectors or self-administration could eliminate this burden [Citation80].

G-CSF agents also pose an additional financial burden to cancer patients beyond their direct cancer care. Biologics (filgrastim, pegfilgrastim, epoetin alfa) in particular tend to be more expensive, and this has limited their accessibility for many patients [Citation22,Citation81]. For example, based on the use of 60 doses per year, the annual effective plan per-patient drug cost is estimated to be approximately 19,000 USD for filgrastim 300-mcg syringes, based on wholesale acquisition drug costs from 2016 [Citation82]. The approval of G-CSF biosimilars, which tend to be less expensive, may alleviate these issues for some patients [Citation81]. In both the US and European G5 countries, cost-efficiency analyses have revealed that biosimilar filgrastim provides significant cost savings relative to originator filgrastim and pegfilgrastim [Citation83–85]. According to modeling of US data, the savings afforded by administration of biosimilar filgrastim could expand access to additional therapeutic or supportive prophylactic care on a budget-neutral basis, thereby increasing access to cancer care to more patients at no additional costs [Citation85].

Given the inherently greater benefit-risk ratio of G-CSF administration, patients who are at a higher risk of developing CIN need to be identified prior to chemotherapy initiation and should receive G-CSF prophylactically. The European Organization for Research and Treatment of Cancer (EORTC), ASCO, and NCCN guidelines, as well as expert guidelines, recommend that patients who have an approximately 20% or higher risk for febrile neutropenia should be administered primary prophylaxis with a G-CSF agent starting with the first cycle of chemotherapy and continuing treatment through subsequent cycles of chemotherapy (). Initial assessment is based on the frequency of febrile neutropenia associated with a patient’s therapeutic regimen. Additional patient- and disease-specific factors are also considered when assessing the potential risk of CIN development (). Patients should be reevaluated for the risk of neutropenia before each subsequent cycle of chemotherapy [Citation12,Citation22,Citation23,Citation68]. This dynamic assessment is supported by results from the MONITOR-GSCF study that evaluated treatment patterns and outcomes of patients treated with chemotherapy and biosimilar filgrastim across 140 global cancer centers and including 1447 patients [Citation86,Citation87]. Physicians can reassess patients based on specific cycle-level risk factors associated with the development of CIN in later cycles, such as the development of CIN in a prior cycle, concomitant antibiotic prophylaxis, impaired performance status, and under-prophylaxis [Citation86]. Accordingly, guidelines recommend that G-CSF should be administered as secondary prophylaxis for patients who did not receive G-CSF but developed febrile neutropenia or a dose-limiting neutropenic event during a prior cycle of chemotherapy [Citation12,Citation22,Citation23,Citation68].

Figure 1. Patient assessment algorithm to determine G-CSF use based on ASCO, EORTC, and NCCN guidelines [Citation12,Citation21,Citation22]. Initial assessment is based on disease and type of chemotherapy regimen. Further refinement of intermediate febrile neutropenia risk is based on patient-specific factors. If indicated, administer G-CSF in the first cycle 24 hours or 3-4 days after completion of myelosuppressive chemotherapy until neutrophil recovery. Patients should be reevaluated prior to second and subsequent chemotherapy cycles. G-CSF should be used as secondary prophylaxis in patients who develop febrile neutropenia or a dose-limiting neutropenic event. In patients who had prior G-CSF, consider chemotherapy dose reduction or change in treatment regimen

a In general, dose-dense regimens require G-CSF support to maintain dose intensity and schedule. b Included in ASCO guidelines. cIncluded in EORTC guidelines. d Included in NCCN Guidelines.ASCO, American Society of Clinical Oncology; EORTC, European Organization for Research and Treatment of Cancer; FN, febrile neutropenia; G-CSF, granulocyte-colony stimulating factor; HIV, human immunodeficiency virus; NCCN, National Comprehensive Cancer Network.
Figure 1. Patient assessment algorithm to determine G-CSF use based on ASCO, EORTC, and NCCN guidelines [Citation12,Citation21,Citation22]. Initial assessment is based on disease and type of chemotherapy regimen. Further refinement of intermediate febrile neutropenia risk is based on patient-specific factors. If indicated, administer G-CSF in the first cycle 24 hours or 3-4 days after completion of myelosuppressive chemotherapy until neutrophil recovery. Patients should be reevaluated prior to second and subsequent chemotherapy cycles. G-CSF should be used as secondary prophylaxis in patients who develop febrile neutropenia or a dose-limiting neutropenic event. In patients who had prior G-CSF, consider chemotherapy dose reduction or change in treatment regimen

Despite these recommendations, these guidelines are often not followed and G-CSF agents are commonly misused. A prospective analysis of 3,638 solid tumor and lymphoma patients from 2002‒2006 reported that prophylactic G-CSF use more than doubled from cycle 1 to cycle 4, while the majority of neutropenic and infection events occurred in cycle 1, suggesting that G-CSF was being used in response to first-cycle neutropenic events rather than as a prophylactic agent in combination with high-risk chemotherapy regimens [Citation8]. A more recent retrospective analysis of 22,868 solid tumor and lymphoma patients also reported under- and mistimed use of G-CSF agents. Of patients with a high risk of developing febrile neutropenia, 76% received prophylactic G-CSF, while only 28% of intermediate-risk patients received G-CSF prophylaxis. Importantly, there was no difference among G-CSF use in intermediate-risk patients who did or did not have ≥1 risk factor for febrile neutropenia, suggesting a lack of awareness of guideline recommendations. Additionally, a concerning proportion of patients (9%) received G-CSF prophylaxis on the same day as completion of chemotherapy rather than the day after chemotherapy as recommended [Citation80]. While data exist that both support and oppose same-day dosing of pegfilgrastim, NCCN guidelines continue to recommend FDA approved dosing schedules of 24 hours or up to 3–4 days post-chemotherapy [Citation22,Citation88,Citation89]. Several studies have also revealed that patients often do not receive the recommended number of doses of filgrastim prophylaxis likely due to perceived patient inconvenience with potential lack of adherence. This practice could result in compromised efficacy [Citation68,Citation90–93]. As a result, expert guidelines state that if there is a risk that filgrastim may not be continued for 11 days, pegfilgrastim may be a preferred option over filgrastim [Citation68]. In summary, these combined observations suggest the importance of continued physician education and awareness of recommended guidelines for G-CSF administration.

5. Emerging agents for prevention and treatment of CIN

Although G-CSF agents have been the standard of care for prevention and treatment of CIN for approximately 30 years, emerging novel agents in various phases of development and approval may help address some of the challenges associated with G-CSF therapy and may change the treatment landscape for CIN () [Citation25].

Table 2. Emerging agents in development to prevent or treat neutropenia

5.1. Eflapegrastim: spectrum pharmaceuticals

Eflapegrastim is a long-acting hematopoietic growth factor consisting of a rhG-CSF analog conjugated via a short polyethylene glycol linker to a human immunoglobulin (IgG) Fc fragment, which extends the half-life of the drug [Citation94]. Eflapegrastim differs from other rhG-CSF agents in that the Fc fragment is able to bind to a receptor called FcRn found on endothelial cells and in the bone marrow, which allows the drug to be retained longer in these tissues [Citation95] . In data from two separate phase 3 clinical trials in early stage breast cancer in 237 and 406 patients treated with TC, eflapegrastim demonstrated noninferiority and comparable safety at a lower G-CSF dose than pegfilgrastim. Consistent with approved G-CSF agents, bone pain remained the most common adverse event associated with eflapegrastim in both studies (all grade ~30%). Other common adverse events across the trials were arthralgia, back pain, and myalgia [Citation94,Citation96]. While these studies were not designed to demonstrate superiority, a pooled analysis of the studies revealed that eflapegrastim significantly reduced the risk of severe neutropenia when compared to pegfilgrastim; however, additional trials are needed to confirm these findings [Citation97]. Ongoing studies are further investigating the CIN-preventative properties of eflapegrastim, including same-day dosing as TC in a phase 1 study in breast cancer patients (NCT04187898), and in a phase 2 study in pediatric patients with solid tumors or lymphomas treated with myelosuppressive therapy (NCT04570423) Clinicaltrials.gov; [Citation98] Available from:https://clinicaltrials.gov/.

5.2. Mecapegfilgrastim: Jiangsu Hengrui Medicine Co., Ltd

Mecapegfilgrastim is a long-acting biosimilar of pegfilgrastim that has shown noninferiority versus short-acting G-CSF agents in phase 3 randomized trials of 339 breast cancer patients treated with anthracycline/taxane or AC and 151 NSCLC patients treated with platinum-based chemotherapy. In both trials, no differences in safety signals were observed among the treatment arms. A statistically significant improvement was observed in the incidence of grade 3/4 neutropenia and the duration of grade 4 neutropenia for mecapegfilgrastim in both trials, suggesting that longer-acting G-CSF may provide clinical benefits to patients; however, additional studies are needed to confirm these findings [Citation99,Citation100]. Mecapegfilgrastim has been approved in China to reduce the incidence of infection in patients with non-myeloid malignancies receiving chemotherapy [Citation101]. It is being further investigated in approximately 20 ongoing phase 2‒4 studies in solid tumors and lymphoma in adults and children in China Clinicaltrials.gov; [Citation98].

5.3. Pegteograstim: Green Cross

Pegteograstim is a long-acting, novel formulation of pegylated rhG-CSF, with a higher biological activity than pegfilgrastim [Citation102]. Pegteograstim has shown promising activity as a prophylactic agent for neutropenia in phase 2 and 3 clinical studies [Citation102,Citation103]. In a randomized noninferiority phase 2/3 study in breast cancer patients (n = 60 and n = 117 for the phase 2 and 3 portions, respectively) treated with docetaxel/doxorubicin (DA) or TAC, pegteograstim was as effective as pegfilgrastim at reducing grade 4 neutropenia in chemotherapy cycle 1 and demonstrated a statistically significantly shorter time to neutrophil recovery. The incidence rates and types of adverse events were similar between the treatment arms [Citation103]. Pegteograstim has been approved to treat neutropenia in South Korea [Citation104].

5.4. Efbemalenograstim alpha (F-627): Evive Biotech

Efbemalenograstim alpha is a long-acting recombinant fusion protein of G-CSF and the Fc domain of human IgG2 that allows once-per-cycle administration to prevent neutropenia [Citation105]. Positive results were observed in a phase 3 trial of efbemalenograstim alpha versus placebo in 122 patients with breast cancer treated with DA. Efbemalenograstim alpha significantly reduced the duration of grade 4 neutropenia in cycle 1 and resulted in a lower incidence rate and duration of grade ≥2 neutropenia and febrile neutropenia. Efbemalenograstim alpha was well tolerated, with the most common treatment-emergent adverse events with a frequency ≥2% higher than placebo being leukopenia, anemia, thrombocytopenia, nausea, and alopecia [Citation106]. Furthermore, a recent pivotal phase 3 clinical study in women with stage I‒III breast cancer treated with TC plus efbemalenograstim alpha or pegfilgrastim met its primary and secondary endpoints, demonstrating that efbemalenograstim alpha was able to prevent CIN [Citation105].

5.5. Plinabulin: BeyondSpring, Inc

Plinabulin is a small molecule that binds to β-tubulin and prevents microtubule polymerization. It is being investigated as both an anti-tumor agent and a neutropenia-preventative agent. Plinabulin has demonstrated anti-tumor activity in vitro against several human tumor cell lines through the induction of apoptosis and anti-angiogenic effects [Citation107–110]. Preclinical studies have also revealed that plinabulin prevents the development of CIN in rodents but does not affect the bone marrow or blood G-CSF levels [Citation111]. These findings have been confirmed in clinical studies, with noninferiority observed versus pegfilgrastim in the number of days of severe neutropenia in a phase 2 randomized study of plinabulin versus pegfilgrastim in 55 NSCLC patients treated with docetaxel [Citation112]. Additionally, results of the phase 3 PROTECTIVE-2 study of plinabulin plus pegfilgrastim compared to pegfilgrastim alone demonstrated that the plinabulin combination results in a significant reduction in the incidence of grade 4 neutropenia, febrile neutropenia, and the duration of profound and severe neutropenia in cycle 1 during TAC treatment in 211 randomized breast cancer patients [Citation113]. The combination regimen also increased the percentage of patients achieving RDI >85% [Citation114]. The safety profile of plinabulin appears manageable. In a phase 1 study in 15 NSCLC patients treated with docetaxel plus plinabulin, the most common adverse events were fatigue, pain, nausea, diarrhea, and vomiting [Citation115]. Furthermore, less bone pain and fewer grade 4 adverse events occurred with plinabulin plus pegfilgrastim versus pegfilgrastim alone in the PROTECTIVE-2 study, and less bone pain was also observed with plinabulin versus pegfilgrastim in the phase 2 study highlighted above in NSCLC [Citation112,Citation113].

Plinabulin has received breakthrough therapy designation for the treatment of CIN by the United States Food & Drug Administration (U.S FDA) and China’s National Medical Products Administration [Citation113]. Plinabulin is currently being investigated as a CIN-preventative agent versus pegfilgrastim in a phase 3 study in NSCLC patients receiving docetaxel (NCT03102606), and as both an anti-cancer agent and CIN-preventative agent in a phase 3 study in advanced NSCLC patients treated with docetaxel (NCT02504489) [Citation98].

5.6. Trilaciclib: G1 Therapeutics, Inc

Similar to plinabulin, trilaciclib is also being investigated as both a CIN-preventative agent and an anti-cancer agent. Trilaciclib is a small-molecule inhibitor of cyclin-dependent kinase 4/6 that is administered intravenously. Preclinically, trilaciclib coadministration with cytotoxic chemotherapy protects murine hematopoietic stem cells from chemotherapy-induced exhaustion by causing transient and reversible G1 cell cycle arrest, resulting in a faster peripheral blood count recovery, enhanced serial transplantation capacity, and reduced myeloid skewing [Citation116]. These benefits have been confirmed in clinical trials. Phase 2 randomized, placebo-controlled clinical studies in extensive-stage small cell lung cancer (SCLC) revealed that trilaciclib administration prior to chemotherapy significantly reduced the duration of severe neutropenia in cycle 1 and the occurrence of severe neutropenia during carboplatin/etoposide, carboplatin/etoposide/atezolizumab, or topotecan treatment (77, 107 and 61 randomized patients, respectively). Improvements in red blood cell and platelet counts, and health-related quality-of-life were also observed [Citation117–119]. Despite these positive results in SCLC, a phase 2 study in 102 randomized TNBC patients treated with gemcitabine/carboplatin revealed that trilaciclib administration did not prevent myelosuppression in this patient population; however, improvements in PFS and OS were observed in the trilaciclib group [Citation120,Citation121]. Across the SCLC trials, trilaciclib appeared to be fairly well-tolerated with no reports of bone pain and fewer grade 3 or higher adverse events than placebo-treated groups, primarily due to fewer hematologic toxicities [Citation117–119,Citation122]

Trilaciclib was approved the US. FDA in February 2021 to reduce the incidence of chemotherapy-induced myelosuppression in adult patients when administered prior to a platinum/etoposide-containing regimen or topotecan-containing regimen for extensive-stage SCLC [Citation123]. Additional ongoing studies are investigating trilaciclib as an anti-cancer agent and a CIN-preventative agent in two phases 3 studies in metastatic colorectal cancer patients treated with FOLFOXIRI/bevacizumab (PRESERVE1; NCT04607668) and metastatic TNBC patients treated with gemcitabine and carboplatin (PRESERVE2; NCT04799249), and a phase 2 study in metastatic NSCLC patients treated with docetaxel (PRESERVE4; NCT04863248).

5.7. Myelo001: Myelo Therapeutics, Inc

Myelo001 is an oral, small molecule that induces differentiation of immature myeloid precursors and has shown CIN-preventative properties in preclinical studies in mice [Citation124]. Data from a randomized phase 2 study of myelo001 versus placebo in 130 breast cancer patients treated with epirubicin/cyclophosphamide suggest that myelo001 may be effective at preventing CIN, though additional follow-up data are needed to confirm these findings because of batch variations noted within the study [Citation125]. Myelo001 is also being investigated for the prevention and treatment of acute radiation syndrome [Citation126].

5.8. EC-18: Enzychem Lifesciences Corporation

EC-18 is a synthetic, orally available, lipid-based small molecule that prevents CIN by attenuating neutrophil extravasation by down-regulation of adhesion molecules, cytokines, and chemokines [Citation127]. In preclinical studies, EC-18 reduced fluorouracil-induced CIN in a mouse model [Citation128], and preliminary results from a phase 1/2 study in metastatic breast cancer suggest that EC-18 combined with AC, has a tolerable safety profile and may prevent CIN [Citation129].

5.9. Romyelocel-L (CTL-008): Cellerant Therapeutics, Inc

Romyelocel-L is a cryopreserved, off-the-shelf human allogeneic myeloid progenitor cell therapeutic manufactured by ex vivo expansion of CD34+ cells isolated from G-CSF–mobilized peripheral blood from healthy donors. The cells are intended to engraft transiently and produce effector cells that migrate to tissues damaged by chemotherapy. In a randomized phase 2 study in 160 acute myeloid leukemia patients treated with standard 7 + 3 induction/consolidation or high-dose cytarabine chemotherapy, romyelocel-L combined with G-CSF reduced infections, antibiotics use, and the length of hospitalization when compared to G-CSF use alone. The safety profile was generally similar between the two arms, including an equal incidence of febrile neutropenia [Citation130].

6. Conclusions

Studies have shown the importance of maintaining RDI to improve overall and disease-free survival, including in elderly and obese patient populations. However, CIN often leads to chemotherapy-dose delays and reductions, subsequently adversely impacting RDI and patient outcomes [Citation7–11]. G-CSF is a well-established prophylactic agent that is recommended by guidelines to reduce neutropenia/febrile neutropenia and improve RDI [Citation12,Citation22,Citation23]. Although G-CSF has been shown to significantly reduce the risk of CIN in those at high risk, there remain several limitations to its use, including side effects and cost [Citation15,Citation22,Citation30,Citation80,Citation81]. Therefore, there is a need to develop and evaluate additional therapies to prevent and manage CIN, which would likely improve RDI and patient outcomes, while simultaneously limiting the potential side effects associated with G-CSF.

7. Expert opinion

Reduced RDI of chemotherapy may be associated with poor outcomes in patients with solid tumors and NHL. CIN is the primary inciting adverse event leading to dose reductions, treatment delays and early treatment cessation, culminating in low RDI. Furthermore, CIN is associated with increased risk of infection, mortality, hospitalizations, and greater financial burdens [Citation2]. We believe it is critical that clinicians identify those patients who are at a high-risk of developing CIN and administer prophylactic G-CSF in a timely and appropriate manner in order to maintain RDI and improve patient outcomes.

Despite the increased use of G-CSF prophylaxis throughout the past decade, many physicians are still not administering G-CSF according to evidence-based recommendations [Citation8,Citation80]. Physicians need to be aware of the importance of maintaining RDI and how to administer G-CSF optimally in their clinical practice. Reasons that may explain deviations from recommended practice include not identifying patients at high risk of CIN, and concerns about G-CSF side effects and costs.

Changing practice patterns for G-CSF administration has also arisen due to the COVID-19 pandemic. In order to reduce the need for hospitalizations and frequent visits to outpatient centers, NCCN provided short-term recommendations specific to COVID-19 for neutropenia-related complications. NCCN recommended expanded prophylactic G-CSF to intermediate-risk patients and therapeutic use to all patients if previously not on G-CSF and febrile neutropenia occurred on a prior cycle. NCCN also has recommended self-administration of G-CSF to avoid frequent visits to outpatient centers [Citation131]. As COVID-19 infections and hospitalizations decrease, how these short-term recommendations will impact long-term clinical practice have yet to be determined. An additional practice pattern that has been occurring is the increased administration of filgrastim over single-injection pegfilgrastim. Anecdotal evidence suggests that U.S. payers are increasingly denying prophylaxis with pegfilgratim and authorizing up to 7 days of daily filgrastim instead due to potential cost savings [Citation84]. While many patients prefer the convenience of once-per-cycle administration [Citation132], how potential changing patterns of U.S. payer approvals will impact future practice is currently unknown. Importantly, these cost savings of daily filgrastim biosimilars over standard filgrastim and pegfilgrastim [Citation83–85] may support the wide-spread use of filgrastim biosimilars as a preferred prophylactic and treatment option for developing countries to increase access to cancer care.

The proper recognition of CIN risk is critically important in order to administer G-CSF in a timely manner. Clinicians need to adequately communicate with patients and their caregivers about the signs and symptoms of CIN and its associated risks and encourage them to immediately report if they develop concerning symptoms to receive prompt and proper management. Regular monitoring and early detection of reduced absolute neutrophil count levels is also critically important to prevent complications of neutropenia.

Several available guidelines and risk models can help physicians identify patients at high-risk of developing CIN [Citation12,Citation22,Citation23]. However, providers may be unsure how to manage patients who are characterized as intermediate-risk for the development of CIN. These patients may not receive prophylactic G-CSF and may then experience febrile neutropenia during their first cycle of treatment. This may result in serious consequences including hospitalization and even death. Subsequent chemotherapy cycles may be delayed or administered at reduced and less effective doses. More research is needed on patients receiving intermediate risk chemotherapy regimens to identify which patients are more likely to develop CIN.

G-CSF products have several limitations that may deter physicians from administering them, including side effects and cost [Citation22,Citation24,Citation81]. One of the reported side effects of G-CSF is bone pain, which can significantly impact patient quality-of-life [Citation22,Citation24]. Furthermore, G-CSF administration is an additional financial burden for patients who are increasingly bearing large portions of the cost of their own care. While the approval of G-CSF biosimilars has some promise of reducing cost moving forward, these agents have a similar side effect profile to first generation G-CSFs [Citation81]. These observations suggest the importance of the development of novel therapies to prevent CIN which may have better safety profiles and lower costs.

Several emerging novel therapies have the potential to change the treatment landscape in the future. Some new drugs represent G-CSF that has been modified to improve availability and decrease administration burden, others have more novel mechanisms of action that may have a greater impact on the treatment landscape used either alone or in combination with G-CSF [Citation25]. Approval of these new agents should be based on randomized-controlled trials demonstrating a reduction in the incidence of neutropenic complications. While we believe that G-CSF will still be widely used as a cancer treatment adjunct in the future, new agents that have both anti-tumor and myeloid-supportive properties look very promising.

Article highlights

  • CIN is the most common adverse event leading to dose alterations and low RDI.

  • The RDI of chemotherapy received is directly associated with patient outcomes.

  • G-CSF prophylaxis is recommended to prevent CIN of select chemotherapy regimens.

  • Novel drugs in development to prevent CIN may change the treatment landscape.

Declaration of interest

M Shayne has served as a consultant for BeyondSpring.

G Lyman’s institution has received funding from Amgen, Lyman has also served as a consultant for G1 Therapeutics, BeyondSpring, Samsung, TEVA and Merck.

DR Harvey’s institution that supports his salary received research funding from Abbisko, AbbVie, Actuate, Alkermes, Amgen, AstraZeneca, Bayer, Bristol-Myers Squibb, Boston Biomedical, Calithera, Fujifilm, Genmab, GlaxoSmithKline, Infinity, lnhibRx, Lycera, Merck, Mersana, Meryx, Nektar, Pfizer, Puma, RAPT Therapeutics, Regeneron, Rgenix, Sanofi, Seattle Genetics, Sutro, Takeda, Xencor. Harvey has also served as a consultant with Amgen and GlaxoSmithKline.

Medical writing support was provided by Rebecca Bigelow (Publication Practice Counsel™; Truposha LLC), which was funded by BeyondSpring, Inc.

Reviewer disclosures

A reviewer on this manuscript holds equity in Matrix45, LLC. Matrix45 has had and/or currently holds contracts with Amgen, Roche, Sandoz/Novartis, Coherus, Teva, Hospira/Pfizer, Mylan (now Viatris).

Reviewers on this manuscript have no other relevant financial relationships or otherwise to disclose.

Correction Statement

This article has been republished with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

This paper received funding from BeyondSpring Inc.

References

  • Hryniuk W, Levine MN. Analysis of dose intensity for adjuvant chemotherapy trials in stage II breast cancer. J Clin Oncol. 1986;4(8):1162–1170.
  • Lalami Y, Klastersky J. Impact of chemotherapy-induced neutropenia (CIN) and febrile neutropenia (FN) on cancer treatment outcomes: an overview about well-established and recently emerging clinical data. Crit Rev Oncol Hematol. 2017;120:163–179.
  • Longo DL, Duffey PL, DeVita VT Jr., et al. The calculation of actual or received dose intensity: a comparison of published methods. J Clin Oncol. 1991;9(11):2042–2051.
  • Bonadonna G, Valagussa P, Moliterni A, et al., Adjuvant cyclophosphamide, methotrexate, and fluorouracil in node-positive breast cancer: the results of 20 years of follow-up. N Engl J Med. 332(14): 901–906. 1995. .
  • Lepage E, Gisselbrecht C, Haioun C, et al., Prognostic significance of received relative dose intensity in non-Hodgkin’s lymphoma patients: application to LNH-87 protocol. The GELA. (Groupe d’Etude des Lymphomes de l’Adulte). Ann Oncol. 4(8): 651–656. 1993.
  • Lyman GH, Dale DC, Crawford J. Incidence and predictors of low dose-intensity in adjuvant breast cancer chemotherapy: a nationwide study of community practices. J Clin Oncol. 2003;21(24):4524–4531.
  • Lyman GH, Dale DC, Friedberg J, et al., Incidence and predictors of low chemotherapy dose-intensity in aggressive non-Hodgkin’s lymphoma: a nationwide study. J Clin Oncol. 22(21): 4302–4311. 2004.
  • Culakova E, Thota R, Poniewierski MS, et al. Patterns of chemotherapy-associated toxicity and supportive care in US oncology practice: a nationwide prospective cohort study. Cancer Med. 2014;3(2):434–444.
  • Kawatkar AA, Farias AJ, Chao C, et al. Hospitalizations, outcomes, and management costs of febrile neutropenia in patients from a managed care population. Support Care Cancer. 2017;25(9):2787–2795.
  • Lyman GH, Dale DC, Tomita D, et al. A retrospective evaluation of chemotherapy dose intensity and supportive care for early-stage breast cancer in a curative setting. Breast Cancer Res Treat. 2013;139(3):863–872.
  • Repetto L. Investigators C. Incidence and clinical impact of chemotherapy induced myelotoxicity in cancer patients: an observational retrospective survey. Crit Rev Oncol Hematol. 2009;72(2):170–179.
  • Aapro MS, Bohlius J, Cameron DA, et al. 2010 update of EORTC guidelines for the use of granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-induced febrile neutropenia in adult patients with lymphoproliferative disorders and solid tumours. Eur J Cancer. 2011;47(1):8–32.
  • Lyman GH, Michels SL, Reynolds MW, et al. Risk of mortality in patients with cancer who experience febrile neutropenia. Cancer. 2010;116(23):5555–5563.
  • Weycker D, Barron R, Kartashov A, et al. Incidence, treatment, and consequences of chemotherapy-induced febrile neutropenia in the inpatient and outpatient settings. J Oncol Pharm Pract. 2014;20(3):190–198.
  • Weycker D, Li X, Tzivelekis S, et al. Burden of chemotherapy-induced febrile neutropenia hospitalizations in US clinical practice, by use and patterns of prophylaxis with colony-stimulating factor. Support Care Cancer. 2017;25(2):439–447.
  • Kuderer NM, Dale DC, Crawford J, et al., Mortality, morbidity, and cost associated with febrile neutropenia in adult cancer patients. Cancer. 106(10): 2258–2266. 2006.
  • Tai E, Jr GPG, Dunbar A, et al. Cost of cancer-related neutropenia or fever hospitalizations, United States, 2012. J Oncol Pract. 2017;13(6):e552–e561. .
  • Clark OA, Lyman GH, Castro AA, et al. Colony-stimulating factors for chemotherapy-induced febrile neutropenia: a meta-analysis of randomized controlled trials. J Clin Oncol. 2005;23(18):4198–4214.
  • Kuderer NM, Dale DC, Crawford J, et al. Impact of primary prophylaxis with granulocyte colony-stimulating factor on febrile neutropenia and mortality in adult cancer patients receiving chemotherapy: a systematic review. J Clin Oncol. 2007;25(21):3158–3167.
  • Lyman GH, Dale DC, Culakova E, et al., The impact of the granulocyte colony-stimulating factor on chemotherapy dose intensity and cancer survival: a systematic review and meta-analysis of randomized controlled trials. Ann Oncol. 24(10): 2475–2484. 2013.
  • Mhaskar R, Clark OA, Lyman G, et al. Colony-stimulating factors for chemotherapy-induced febrile neutropenia. Cochrane Database Syst Re. 2014(10):CD003039
  • Hematopoietic growth factors: version 2. 2021. National Comprehensive Cancer Network; [updated 2021 Mar 23; cited 2021 May 12]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/growthfactors.pdf
  • Smith TJ, Bohlke K, Lyman GH, et al. Recommendations for the use of WBC growth factors: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2015;33(28):3199–3212.
  • D’Souza A, Jaiyesimi I, Trainor L, et al. Granulocyte colony-stimulating factor administration: adverse events. Transfus Med Rev. 2008;22(4):280–290. .
  • Matikas A, Georgoulias V, Kotsakis A. Emerging agents for the prevention of treatment induced neutropenia in adult cancer patients. Expert Opin Emerg Drugs. 2016;21(2):157–166.
  • Shayne M, Crawford J, Dale DC, et al. Predictors of reduced dose intensity in patients with early-stage breast cancer receiving adjuvant chemotherapy. Breast Cancer Res Treat. 2006;100(3):255–262.
  • Culakova E, Poniewierski MS, Wolff DA, et al. The impact of chemotherapy dose intensity and supportive care on the risk of febrile neutropenia in patients with early stage breast cancer: a prospective cohort study. SpringerPlus. 2015;4(1):396.
  • Schwartzberg LS, Saleh M, Whittaker S, et al. Severe neutropenia and relative dose intensity among patients<65 and ≥65 years with non-hodgkin’s lymphoma receiving chop-based chemotherapy. Support Care Cancer. 2014;22(7):1833–1841.
  • Weycker D, Barron R, Edelsberg J, et al. Incidence of reduced chemotherapy relative dose intensity among women with early stage breast cancer in US clinical practice. Breast Cancer Res Treat. 2012;133(1):301–310.
  • Lyman GH, Crawford J, Tomita D, et al. Changing patterns of chemotherapy relative dose intensity and supportive care for aggressive B-cell non-Hodgkin lymphoma. Leuk Lymphoma. 2016;57(2):283–290. .
  • Citron ML. Dose-dense chemotherapy: principles, clinical results and future perspectives. Breast Care (Basel). 2008;3(4):251–255.
  • Gray R, Bradley R, Braybrooke J, Early Breast Cancer Trialists’ Collaborative Group. Increasing the dose intensity of chemotherapy by more frequent administration or sequential scheduling: a patient-level meta-analysis of 37 298 women with early breast cancer in 26 randomised trials. Lancet. 2019;393(10179):1440–1452. .
  • Wood WC, Budman DR, Korzun AH, et al. Dose and dose intensity of adjuvant chemotherapy for stage II, node-positive breast carcinoma. N Engl J Med. 1994;330(18):1253–1259.
  • Choueiri TK, Jacobus S, Bellmunt J, et al. Neoadjuvant dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin with pegfilgrastim support in muscle-invasive urothelial cancer: pathologic, radiologic, and biomarker correlates. J Clin Oncol. 2014;32(18):1889–1894.
  • Culine S, Gravis G, Flechon A, et al. Randomized phase III trial of dose-dense methotrexate, vinblastine, doxorubicin, and cisplatin (dd-MVAC) or gemcitabine and cisplatin (GC) as perioperative chemotherapy for muscle invasive urothelial bladder cancer (MIUBC): preliminary results of the GETUG/AFU V05 VESPER trial on toxicity and pathological responses. J Clin Oncol. 2020;38(6_suppl):437.
  • Plimack ER, Hoffman-Censits JH, Viterbo R, et al. Accelerated methotrexate, vinblastine, doxorubicin, and cisplatin is safe, effective, and efficient neoadjuvant treatment for muscle-invasive bladder cancer: results of a multicenter phase II study with molecular correlates of response and toxicity. J Clin Oncol. 2014;32(18):1895–1901.
  • Havrilesky LJ, Reiner M, Morrow PK, et al. A review of relative dose intensity and survival in patients with metastatic solid tumors. Crit Rev Oncol Hematol. 2015;93(3):203–210.
  • Anuradha S, Donovan PJ, Webb PM, et al. Variations in adjuvant chemotherapy and survival in women with epithelial ovarian cancer - a population-based study. Acta Oncol. 2016;55(2):226–233.
  • Fauci JM, Whitworth JM, Schneider KE, et al. Prognostic significance of the relative dose intensity of chemotherapy in primary treatment of epithelial ovarian cancer. Gynecol Oncol. 2011;122(3):532–535.
  • Bonadonna G, Moliterni A, Zambetti M, et al. 30 years’ follow up of randomised studies of adjuvant CMF in operable breast cancer: cohort study. BMJ. 2005;330(7485):217.
  • Abdel-Rahman O. Outcomes of early-stage breast cancer patients treated with sequential anthracyclines-taxanes in relationship to relative dosing intensity: a secondary analysis of a randomized controlled trial. Clin Transl Oncol. 2019;21(2):239–245.
  • Cespedes Feliciano EM, Chen WY, Lee V, et al. Body composition, adherence to anthracycline and taxane-based chemotherapy, and survival after nonmetastatic breast cancer. JAMA Oncol. 2020;6(2):264–270.
  • Zhang L, Yu Q, Wu XC, et al. Impact of chemotherapy relative dose intensity on cause-specific and overall survival for stage I-III breast cancer: ER+/PR+, HER2- vs. triple-negative. Breast Cancer Res Treat. 2018;169(1):175–187.
  • Aspinall SL, Good CB, Zhao X, et al. Adjuvant chemotherapy for stage III colon cancer: relative dose intensity and survival among veterans. BMC Cancer. 2015;15:62.
  • Van Eeghen EE, Bakker SD, van Bochove A, et al. High risk stage 2 and stage 3 colon cancer, predictors of recurrence and effect of adjuvant therapy in a nonselected population. Int Sch Res Notices. 2015;2015:790186.
  • Yabusaki N, Fujii T, Yamada S, et al. The significance of relative dose intensity in adjuvant chemotherapy of pancreatic ductal adenocarcinoma—including the analysis of clinicopathological factors influencing relative dose intensity. Medicine (Baltimore). 2016;95(29):e4282.
  • Hirakawa T, Yamaguchi H, Yokose N, et al. Importance of maintaining the relative dose intensity of CHOP-like regimens combined with rituximab in patients with diffuse large B-cell lymphoma. Ann Hematol. 2010;89(9):897–904.
  • Lee S, Fujita K, Negoro E, et al. Impact of relative dose intensity of standard regimens on survival in elderly patients aged 80 years and older with diffuse large B-cell lymphoma. Haematologica. 2020;105(8):e415–e418.
  • Dlugosz-Danecka M, Szmit S, Ogorka T, et al. The average relative dose intensity of R-CHOP is an independent factor determining favorable overall survival in diffuse large B-cell lymphoma patients. Cancer Med. 2019;8(3):1103–1109.
  • Yamamoto M, Suzuki I, Saitou K, et al. Impact of comorbidity and relative dose intensity on outcomes in diffuse large B-cell lymphoma patients treated with R-CHOP. J Cancer Res Clin Oncol. 2020;146(11):2995–3002.
  • Gutiérrez A, Bento L, Bautista-Gili AM, et al. Differential impact of relative dose-intensity reductions in diffuse large B-cell lymphoma treated with R-CHOP21 or R-CHOP14. PLoS One. 2015;10(4):e0123978.
  • Klasa RJ, Murray N, Coldman AJ. Dose-intensity meta-analysis of chemotherapy regimens in small-cell carcinoma of the lung. J Clin Oncol. 1991;9(3):499–508.
  • Ladwa R, Kalas T, Pathmanathan S, et al. Maintaining dose intensity of adjuvant chemotherapy in older patients with breast cancer. Clin Breast Cancer. 2018;18(5):e1181–e1187.
  • Schraa SJ, Frerichs KA, Agterof MJ, et al. Relative dose intensity as a proxy measure of quality and prognosis in adjuvant chemotherapy for breast cancer in daily clinical practice. Eur J Cancer. 2017;79:152–157.
  • Smoragiewicz M, Javaheri KR, Yin Y, et al. Neutropenia and relative dose intensity on adjuvant FOLFOX chemotherapy are not associated with survival for resected colon cancer. J Gastrointest Cancer. 2014;45(4):460–465.
  • Sparano JA, Wang M, Martino S, et al. Weekly paclitaxel in the adjuvant treatment of breast cancer. N Engl J Med. 2008;358(16):1663–1671.
  • Shayne M, Culakova E, Poniewierski MS, et al. Dose intensity and hematologic toxicity in older cancer patients receiving systemic chemotherapy. Cancer. 2007;110(7):1611–1620.
  • Raza S, Welch S, Younus J. Relative dose intensity delivered to patients with early breast cancer: canadian experience. Curr Oncol. 2009;16(6):8–12.
  • Li S, Liu J, Guo H, et al. Trends in the use of primary prophylactic colony-stimulating factors and neutropenia-related hospitalization in elderly cancer patients receiving myelosuppressive chemotherapy in the USA: 1995-2015. Support Care Cancer. 2020;28(6):2637–2649.
  • Older adult oncology: verson 1. 2020 February 7. National Comprehensive Cancer Network; [ updated 2020 Nov 10; cited 2020 Jan 5]. Available from: https://www.nccn.org/about/news/ebulletin/ebulletindetail.aspx?ebulletinid=1578
  • Denduluri N, Patt DA, Wang Y, et al. Dose delays, dose reductions, and relative dose intensity in patients with cancer who received adjuvant or neoadjuvant chemotherapy in community oncology practices. J National Compr Cancer Network. 2015;13(11):1383.
  • Griggs JJ, Mangu PB, Anderson H, et al. Appropriate chemotherapy dosing for obese adult patients with cancer: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2012;30(13):1553–1561.
  • Griggs JJ, Sorbero ME, Lyman GH. Undertreatment of obese women receiving breast cancer chemotherapy. Arch Intern Med. 2005;165(11):1267–1273.
  • Meyerhardt JA, Catalano PJ, Haller DG, et al. Influence of body mass index on outcomes and treatment-related toxicity in patients with colon carcinoma. Cancer. 2003;98(3):484–495.
  • Meyerhardt JA, Niedzwiecki D, Hollis D, et al. Impact of body mass index and weight change after treatment on cancer recurrence and survival in patients with stage III colon cancer: findings from Cancer and Leukemia Group B 89803. J Clin Oncol. 2008;26(25):4109–4115.
  • Meyerhardt JA, Tepper JE, Niedzwiecki D, et al. Impact of body mass index on outcomes and treatment-related toxicity in patients with stage II and III rectal cancer: findings from Intergroup Trial 0114. J Clin Oncol. 2004;22(4):648–657.
  • Poikonen P. Effect of obesity on the leukocyte nadir in women treated with adjuvant cyclophosphamide, methotrexate, and fluorouracil dosed according to body surface area. Acta Oncol. 2001;40(1):67–71.
  • Aapro M, Boccia R, Leonard R, et al. Refining the role of pegfilgrastim (a long-acting G-CSF) for prevention of chemotherapy-induced febrile neutropenia: consensus guidance recommendations. Support Care Cancer. 2017;25(11):3295–3304.
  • Awad M, Singh P, Zarxio HO. filgrastim-sndz): the first biosimilar approved by the FDA. P T. 2017;42(1):19–23.
  • Drug approval package: neulasta (pegfilgrastim) injection [Internet]: US Food & Drug Administration; 2002 [ updated Jan 2002; cited 2020 Nov 16]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/125031_0000_NeulastaTOC.cfm
  • Biosimilar and Interchangeable Biologics: more treatment choices [Internet]: US Food and Drug Administration; 2020 [ updated 2020 Mar 23; cited 2021 May 18]. Available from: https://www.fda.gov/consumers/consumer-updates/biosimilar-and-interchangeable-biologics-more-treatment-choices
  • Agboola F, Reddy P. Conversion from Filgrastim to Tbo-filgrastim: experience of a Large Health Care System. J Manag Care Spec Pharm. 2017;23(12):1214–1218.
  • Neupogen (filgrastim) prescribing information [Internet]. Thousand Oaks, CA: Amgen. 2018. updated Jun 2018; cited 2020 Oct 15]. Available from: https://www.pi.amgen.com/~/media/amgen/repositorysites/pi-amgen-com/neupogen/neupogen_pi_hcp_english.pdf
  • Granix (tbo-filgrastim) prescribing information [Internet] North Wales, PA: cephalon; 2019 [ updated Nov 2019; cited 2020 Oct 15]. Available from: https://www.granixhcp.com/globalassets/granix-hcp/prescribing-information.pdf
  • Weycker D, Bensink M, Lonshteyn A, et al. Risk of chemotherapy-induced febrile neutropenia by day of pegfilgrastim prophylaxis in US clinical practice from 2010 to 2015. Curr Med Res Opin. 2017;33(12):2107–2113.
  • Weycker D, Li X, Figueredo J, et al. Risk of chemotherapy-induced febrile neutropenia in cancer patients receiving pegfilgrastim prophylaxis: does timing of administration matter? Support Care Cancer. 2016;24(5):2309–2316.
  • Papakonstantinou A, Hedayati E, Hellstrom M, et al. Neutropenic complications in the PANTHER phase III study of adjuvant tailored dose-dense chemotherapy in early breast cancer. Acta Oncol. 2020;59(1):75–81.
  • Wang L, Baser O, Kutikova L, et al. The impact of primary prophylaxis with granulocyte colony-stimulating factors on febrile neutropenia during chemotherapy: a systematic review and meta-analysis of randomized controlled trials. Support Care Cancer. 2015;23(11):3131–3140.
  • Neulasta (pegfilgrastim) prescribing information [Internet]. Thousand Oaks, CA: Amgen; 2020;[ updated Jan 2020; cited 2020 Oct 15]. Available from: https://www.pi.amgen.com/~/media/amgen/repositorysites/pi-amgen-com/neulasta/neulasta_pi_hcp_english.pdf
  • Gawade PL, Li S, Henry D, et al. Patterns of granulocyte colony-stimulating factor prophylaxis in patients with cancer receiving myelosuppressive chemotherapy. Support Care Cancer. 2020;28(9):4413–4424.
  • Dinan MA, Hirsch BR, Lyman GH. Management of chemotherapy-induced neutropenia: measuring quality, cost, and value. J Natl Compr Canc Netw. 2015;13(1):e1–7.
  • Trautman H, Szabo E, James E, et al. Patient-administered biologic and biosimilar filgrastim may offer more affordable options for patients with nonmyeloid malignancies receiving chemotherapy in the United States: a budget impact analysis from the payer perspective. J Manag Care Spec Pharm. 2019;25(1):94–101.
  • Aapro M, Cornes P, Abraham I. Comparative cost-efficiency across the European G5 countries of various regimens of filgrastim, biosimilar filgrastim, and pegfilgrastim to reduce the incidence of chemotherapy-induced febrile neutropenia. J Oncol Pharm Pract. 2012;18(2):171–179.
  • McBride A, Balu S, Campbell K, et al. Expanded access to cancer treatments from conversion to neutropenia prophylaxis with biosimilar filgrastim-sndz. Future Oncol. 2017;13(25):2285–2295.
  • McBride A, Campbell K, Bikkina M, et al. Cost-efficiency analyses for the US of biosimilar filgrastim-sndz, reference filgrastim, pegfilgrastim, and pegfilgrastim with on-body injector in the prophylaxis of chemotherapy-induced (febrile) neutropenia. J Med Econ. 2017;20(10):1083–1093.
  • Aapro M, Ludwig H, Bokemeyer C, et al. Predictive modeling of the outcomes of chemotherapy-induced (febrile) neutropenia prophylaxis with biosimilar filgrastim (MONITOR-GCSF study). Ann Oncol. 2016;27(11):2039–2045.
  • Gascón P, Aapro M, Ludwig H, et al. Treatment patterns and outcomes in the prophylaxis of chemotherapy-induced (febrile) neutropenia with biosimilar filgrastim (the MONITOR-GCSF study). Support Care Cancer. 2016;24(2):911–925.
  • Eckstrom J, Bartels T, Abraham I, et al. A single-arm, retrospective analysis of the incidence of febrile neutropenia using same-day versus next-day pegfilgrastim in patients with gastrointestinal cancers treated with FOLFOX or FOLFIRI. Support Care Cancer. 2019;27(3):873–878.
  • Lyman GH, Allcott K, Garcia J, et al. The effectiveness and safety of same-day versus next-day administration of long-acting granulocyte colony-stimulating factors for the prophylaxis of chemotherapy-induced neutropenia: a systematic review. Support Care Cancer. 2017;25(8):2619–2629.
  • Weycker D, Hackett J, Edelsberg JS, et al. Are shorter courses of filgrastim prophylaxis associated with increased risk of hospitalization? Ann Pharmacother. 2006;40(3):402–407. .
  • Almenar Cubells D, Bosch Roig C, Jiménez Orozco E, et al. Effectiveness of daily versus non-daily granulocyte colony-stimulating factors in patients with solid tumours undergoing chemotherapy: a multivariate analysis of data from current practice. Eur J Cancer Care (Engl). 2013;22(3):400–412.
  • Almenar D, Mayans J, Juan O, et al. Pegfilgrastim and daily granulocyte colony-stimulating factor: patterns of use and neutropenia-related outcomes in cancer patients in Spain--results of the LEARN Study. Eur J Cancer Care (Engl). 2009;18(3):280–286.
  • Naeim A, Henk HJ, Becker L, et al. Pegfilgrastim prophylaxis is associated with a lower risk of hospitalization of cancer patients than filgrastim prophylaxis: a retrospective United States claims analysis of granulocyte colony-stimulating factors (G-CSF). BMC Cancer. 2013;13:11.
  • Cobb PW, Moon YW, Mezei K, et al. A comparison of eflapegrastim to pegfilgrastim in the management of chemotherapy-induced neutropenia in patients with early-stage breast cancer undergoing cytotoxic chemotherapy (RECOVER): a phase 3 study. Cancer Med. 2020;9(17):6234–6243.
  • Barrett JA, Choi J, Lakshmikanthan S, et al. Eflapegrastim’s enhancement of efficacy compared with pegfilgrastim in neutropenic rats supports potential for same-day dosing. Experimental Hematology. 2020;92:51–61.
  • Schwartzberg LS, Bhat G, Peguero J, et al. Eflapegrastim, a long-acting granulocyte-colony stimulating factor for the management of chemotherapy-induced neutropenia: results of a phase III trial. Oncologist 2020;25. doi:https://doi.org/10.1634/theoncologist.2020-0105.
  • Schwartzberg LS, Bhat G, Restrepo A, et al. Pooled efficacy analysis from two phase 3 studies in patients receiving eflapegrastim, a novel, long-acting granulocyte-colony stimulating factor, following TC for early-stage breast cancer. Poster Presented at the Virtual San Antonio Breast Cancer Symposium. 2020; Abstract PS9–59.
  • Clinicaltrials.gov. 2020. cited 2020 Dec 12]. Available from: https://clinicaltrials.gov/
  • Xu F, Zhang Y, Miao Z, et al. Efficacy and safety of mecapegfilgrastim for prophylaxis of chemotherapy-induced neutropenia in patients with breast cancer: a randomized, multicenter, active-controlled phase III trial. Ann Transl Med. 2019;7(18):482. .
  • Zhou C, Huang Y, Wang D, et al. A randomized multicenter phase iii study of single administration of mecapegfilgrastim (HHPG-19K), a pegfilgrastim biosimilar, for prophylaxis of chemotherapy-induced neutropenia in patients with advanced non-small-cell lung cancer (NSCLC). Clin Lung Cancer. 2016;17(2):119–127.
  • Tirelli U, Carbone A, Di Francia R, et al. A new peg-filgrastim biosimilar, mecapegfilgrastim for primary prophylaxis of chemotherapy-related neutropenia is now available. Ann Transl Med. 2020;8(5):166.
  • Kim GM, Kim JH, Kim JH, et al. A phase II study to evaluate the safety and efficacy of pegteograstim in Korean breast cancer patients receiving dose-dense doxorubicin/cyclophosphamide. Cancer Res Treat. 2019;51(2):812–818.
  • Lee KH, Kim JY, Lee MH, et al. A randomized, multicenter, phase II/III study to determine the optimal dose and to evaluate the efficacy and safety of pegteograstim (GCPGC) on chemotherapy-induced neutropenia compared to pegfilgrastim in breast cancer patients: KCSG PC10-09. Support Care Cancer. 2016;24(4):1709–1717.
  • Pegteograstim biological approved in South Korea [Internet]: 2014. updated September 19, 2014; cited 2020 Nov 19]. Available from:. http://www.gabionline.net/Biosimilars/News/Pegteograstim-biological-approved-in-South-Korea
  • Evive Biotech meets primary and secondary endpoints in global phase III clinical trial for their novel chemotherapy-induced neutropenia treatment [Internet]: Evive Biotech; 2020 [ updated 2020 Jul 7; cited 2020 Dec 14]. Available from: https://www.evivebiotech.com/press_releases/evive-biotech-meets-primary-and-secondary-endpoints-in-global-phase-iii-clinical-trial-for-their-novel-chemotherapy-induced-neutropenia-treatment/
  • Glaspy J, Tang T, Rutty D, et al. A phase III, randomized, multi-centre, double-blind, placebo-controlled clinical trial of F-627 in women with breast cancer receiving myelotoxic chemotherapy. 2018 San Antonio Breast Cancer Symposium; December 4–8, 2018 2018: abstract P6-13–03. San Antonio, TX.
  • Nicholson B, Lloyd GK, Miller BR, et al. NPI-2358 is a tubulin-depolymerizing agent: in-vitro evidence for activity as a tumor vascular-disrupting agent. Anticancer Drugs. 2006;17(1):25–31.
  • Kashyap AS, Fernandez-Rodriguez L, Zhao Y, et al. GEF-H1 signaling upon microtubule destabilization is required for dendritic cell activation and specific anti-tumor responses. Cell Rep. 2019;28(13):3367–3380 e8.
  • Singh AV, Bandi M, Raje N, et al. A novel vascular disrupting agent plinabulin triggers JNK-mediated apoptosis and inhibits angiogenesis in multiple myeloma cells. Blood. 2011;117(21):5692–5700.
  • Wang Y, Zhang H, Gigant B, et al. Structures of a diverse set of colchicine binding site inhibitors in complex with tubulin provide a rationale for drug discovery. FEBS J. 2016;283(1):102–111.
  • Tonra JR, Lloyd GK, Mohanlal R, et al. Plinabulin ameliorates neutropenia induced by multiple chemotherapies through a mechanism distinct from G-CSF therapies. Cancer Chemother Pharmacol. 2020;85(2):461–468.
  • Blayney DW, Zhang Q, Feng J, et al. Efficacy of plinabulin vs pegfilgrastim for prevention of chemotherapy-induced neutropenia in adults with non-small cell lung cancer: a phase 2 randomized clinical trial. JAMA Oncol. 2020;6:e204429.
  • BeyondSpring announces positive topline results from its PROTECTIVE-2 phase 3 registrational trial of plinabulin in combination with pegfilgrastim for prevention of chemotherapy-induced neutropenia [Internet]: BeyondSpring; 2020 [ cited 2020 Dec 12]. Available from: https://www.beyondspringpharma.com/pressreleases/info.aspx?itemid=4239
  • Blayney DW, Ginn G, Huang L, et al. Plinabulin and pegfilgrastim (Plin+Peg) versus peg monotherapy (Peg) after TAC: a comparison of efficacy, safety, relative dose intensity (RDI) and bone pain. Poster Presented at the Virtual San Antonio Breast Cancer Symposium 2020; Abstract PS11–15.
  • Millward M, Mainwaring P, Mita A, et al. Phase 1 study of the novel vascular disrupting agent plinabulin (NPI-2358) and docetaxel. Invest New Drugs. 2012;30(3):1065–1073.
  • He S, Roberts PJ, Sorrentino JA, et al. Transient CDK4/6 inhibition protects hematopoietic stem cells from chemotherapy-induced exhaustion. Sci Transl Med. 2017;9:387.
  • Daniel D, Kuchava V, Bondarenko I, et al. Trilaciclib prior to chemotherapy and atezolizumab in patients with newly diagnosed extensive-stage small cell lung cancer: a multicentre, randomised, double-blind, placebo-controlled phase II trial. Int J Cancer. 2020. https://doi.org/10.1002/ijc.33453.
  • Hart LL, Ferrarotto R, Andric ZG, et al. Myelopreservation with Trilaciclib in patients receiving topotecan for small cell lung cancer: results from a randomized, double-blind, placebo-controlled phase II study. Adv Ther. 2021;38(1):350–365.
  • Weiss JM, Csoszi T, Maglakelidze M, et al. Myelopreservation with the CDK4/6 inhibitor trilaciclib in patients with small-cell lung cancer receiving first-line chemotherapy: a phase Ib/randomized phase II trial. Ann Oncol. 2019;30(10):1613–1621.
  • O’Shaughnessy J, Wright G, Thummala A, et al. Trilaciclib improves overall survival when given with gemcitabine/carboplatin in patients with metastatic triple-negative breast cancer: final analysis of a randomized phase 2 trial. Presented at the 2020 Virtual Meeting of the San Antonio Breast Cancer Symposium December 8-11, 2020 Abstract PD1–06.
  • Tan AR, Wright GS, Thummala AR, et al. Trilaciclib plus chemotherapy versus chemotherapy alone in patients with metastatic triple-negative breast cancer: a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol. 2019;20(11):1587–1601.
  • Weiss J, Goldschmidt J, Zoran A, et al. Myelopreservation and reduced use of supportive care with trilaciclib in patients with small cell lung cancer. J Clin Oncol. 2020;38(15_suppl):12096.
  • FDA approves drug to reduce bone marrow suppression caused by chemotherapy [Internet]: U.S. Food & Drug Administration; 2021 [ updated 2021 Feb 12; cited 2021 May 10]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-drug-reduce-bone-marrow-suppression-caused-chemotherapy
  • Pleimes D, Flechsig S, Meyer M. Effect of IEPA, a novel orally bioavailable small molecule, on chemotherapy-induced myelosuppression. Blood. 2014;124(21):4355.
  • Mayer F, Pleimes D, Schmidt M, et al. Investigation of the efficacy of imidazolyl ethanamide pentandioic acid (IEPA) to reduce chemotherapy-induced myelosuppression (CIM) in breast cancer patients. J Clin Oncol. 2019;37(15_suppl):e23098–e23098.
  • Pleimes D, Bunker V, Meyer M, et al. Effects of a novel orally bioavailable small molecule (imidazolyl ethanamide pentandioic acid) on acute radiation syndrome. Blood. 2018;132(Supplement 1):5089.
  • Jeong J, Kim Y-J, Lee DY, et al. 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation. Cell Biosci. 2019;9(1):4.
  • Kim Y-J, Jeong J, Sohn K-Y, et al. Abstract 360: therapeutic potential of EC-18 as a chemotherapy adjuvant for 5-fluorouracil-induced neutropenia. Cancer Re. 2019;79(13 Supplement):360.
  • Kim S-B, Kim M-H. Abstract A070: a phase 1/2a, open-label, dose-escalation study of EC-18 in patients with metastatic breast cancer for the prevention of chemotherapy-induced neutropenia. Mol Cancer Ther. 2019;18(12 Supplement):A070.
  • Abboud CN, Akard LP, Andreadis C, et al. A randomized open label exploratory controlled trial of CLT-008 myeloid progenitor cells (MPC) to decrease infections during induction for AML. J Clin Oncol. 2018;36(15_suppl):7043.
  • Griffiths EA, Alwan LM, Bachiashvili K, et al. Considerations for use of hematopoietic growth factors in patients with cancer related to the COVID-19 pandemic. J Natl Compr Canc Netw 2020 1–4 doi:https://doi.org/10.6004/jnccn.2020.7610
  • Johnson P, Bancroft T, Barron R, et al. Discrete choice experiment to estimate breast cancer patients’ preferences and willingness to pay for prophylactic granulocyte colony-stimulating factors. Value Health. 2014;17(4):380–389.