We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

The impact of myelosuppression on quality of life of patients treated with chemotherapy

    Jeffrey Crawford

    *Author for correspondence:

    E-mail Address: jeffrey.a.crawford@duke.edu

    Duke Cancer Institute, Duke Medicine, Durham, NC 27710, USA

    ,
    Dana Herndon

    Cone Health Cancer Center, Greensboro, NC 27403, USA

    ,
    Katerina Gmitter

    Patient Author, Chapel Hill, NC 27516, USA

    &
    Jared Weiss

    UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA

    Published Online:https://doi.org/10.2217/fon-2023-0513

    Abstract

    Side effects from chemotherapy-induced myelosuppression can negatively affect patients' quality of life (QoL). Neutropenia increases infection risk, and anemia frequently results in debilitating fatigue. Additionally, the bleeding risk associated with thrombocytopenia can lead to fear and anxiety. However, traditional interventions for myelosuppression fall short of the ideal. Granulocyte colony-stimulating factors reduce the risk of severe neutropenia but commonly lead to bone pain. Erythropoiesis-stimulating agents are not always effective and may cause thromboembolic events, while transfusions to correct anemia/thrombocytopenia are associated with transfusion reactions and volume overload. Trilaciclib, which is approved for reducing myelosuppression in patients with extensive-stage small cell lung cancer, together with several investigational agents in development for managing myelosuppression have the potential to improve QoL for patients on chemotherapy.

    Plain language summary

    Chemotherapy can cause side effects by killing blood-forming cells in the bone marrow. This is known as myelosuppression and leads to neutropenia (decreased neutrophils [white blood cells]), anemia (decreased red blood cells) and thrombocytopenia (decreased platelets). Neutropenia can increase the risk of getting an infection, and severe cases might result in patients being hospitalized. Both neutropenia and anemia can cause fatigue, which is often reported by patients as being the most draining symptom of chemotherapy. Thrombocytopenia increases the risk of bleeding and can cause patients with cancer to become even more scared and anxious. Myelosuppression due to chemotherapy is usually managed with delays or reductions in the amount of chemotherapy that patients receive, but this may worsen the disease. Other treatments, known as supportive care interventions, include growth factors, which stimulate the production of blood cells and red blood cell or platelet transfusions. However, having these treatments in addition to chemotherapy can be a burden to patients, and they can cause side effects such as bone pain and blood clots. A treatment called trilaciclib is approved by the US Food and Drug Administration for patients receiving certain types of chemotherapy for advanced small-cell lung cancer. Trilaciclib has been shown to reduce neutropenia, anemia and thrombocytopenia in these patients and improve their quality of life. Other drugs are also being assessed in clinical trials for preventing or treating myelosuppression in patients with different cancer types. In the future, these drugs may improve quality of life for patients on chemotherapy.

    Tweetable abstract

    The burden of chemotherapy-induced myelosuppression on patients with cancer is substantial. We provide insights on the impact of this side effect on the lives of patients and review management strategies aimed at improving outcomes.

    Despite the emergence of targeted drugs and recent advances in immunotherapy, chemotherapy remains a cornerstone of treatment for many people with cancer [1]. Unfortunately, the side effects of chemotherapy can have a negative impact on quality of life (QoL) [1,2].

    Chemotherapies target not only cancer cells, but also healthy cells that divide quickly, including hair cells and cells in the digestive system, which can result in hair loss (alopecia), nausea, vomiting and diarrhea. Chemotherapy can also kill blood-forming cells known as hematopoietic stem and progenitor cells (HSPCs) found in the bone marrow [3]. HSPCs give rise to normal blood cells, including white blood cells (WBCs), red blood cells (RBCs) and platelets. Destruction of HSPCs, known as myelosuppression, can reduce the numbers of one, two or all of these blood cell types, resulting in neutropenia (decreased neutrophils, which are a type of WBC), anemia (decreased RBCs), thrombocytopenia (decreased platelets) and/or pancytopenia (decrease in all three types). The presentation of myelosuppression and how often it occurs depends on the individual person, the chemotherapy that is used and when it is administered [3,4].

    The symptoms of chemotherapy-induced myelosuppression can have a negative impact on patients' QoL [1,2]. In Box 1, we present insights from our patient author, Katerina Gmitter, in which she describes the numerous side effects of chemotherapy treatment for triple-negative breast cancer, how they impacted her life, and the need for medical professionals to proactively discuss the side effects of chemotherapy with each patient.

    Box 1. Patient insights.

    Patient background:

    Katerina is a survivor of triple-negative breast cancer (TNBC), having been diagnosed in 2010, at the age of 32 years. Her two children were aged 4 months and 2.5 years at the time of the diagnosis.

    Katerina was 20 years old when her mother died from metastatic TNBC at the age of 48 years. Katerina and her two sisters are carriers of mutated BRCA genes. Katerina's younger sister was also subsequently diagnosed with TNBC and went through surgery and chemotherapy, while her other sister had prophylactic surgery.

    Treatment received:

    Katerina underwent bilateral mastectomy surgery within 2 weeks of her TNBC diagnosis. Chemotherapy treatment was with adriamycin, cytoxan and taxol. Pegfilgrastim was given with every chemotherapy cycle, but she did not require any antibiotic treatments or blood transfusions. Katerina received chemotherapy for 6 months and bevacizumab for 1 year as part of a clinical trial.

    Side effects of chemotherapy & their effects on quality of life:

    Chemotherapy, for me, was worse than anything that I could have ever expected. I faced extreme brain fog, exhaustion, migraines, dehydration, mouth sores, black and painful nails that eventually fell off, neutropenia, vaginal dryness (having sex was impossible), chronic constipation, neuropathy, extreme nausea to the point of needing intravenous (iv.) fluids and anxiety.

    I had anticipatory post-traumatic stress disorder responses to the adriamycin. As soon as I walked through the doors of where I received chemotherapy, I would start to feel nauseous. When I sat in the chair and saw the nurse coming toward me with the red syringe, I would start dry-heaving and vomiting.

    Chemotherapy, in general, is depleting and exhausting and, for me, it was completely life changing. After the first round of chemotherapy, I arrived home, and was on the bathroom floor the entire night. Not knowing what the next day would look like, I would just try to take it one day at a time, one moment at a time, sometimes just one breath at a time, trying to make it through.

    My life as I knew it was put on hold, and I could not make plans because I never knew how I would feel. I needed someone with me to care for my children: I was too fatigued and sick to give them the care they deserved. I was so exhausted that there were days I could not get out of bed. I would try to at least move and walk, but there were days I could not even make it down the block from exhaustion and fatigue.

    One of the side effects that I never would have thought would impact my life so much was my nails – the fact that they were so painful. When I tried to spend time with my kids, when my son was 2.5 and then 3 years old, he would accidentally step on my toe and it was just excruciating pain, so I could not be barefoot, but shoes would hurt. These little things really do have a great impact.

    At age 32, my body went into menopause due to my chemotherapy regimen, forever altering my bone, heart and brain health. I developed chemical sensitivities following my chemotherapy regimen.

    The worst side effects were fatigue, constipation and nausea. If I were not so fatigued, I think the other two would have been easier to tolerate.

    Impact & treatment of neutropenia:

    The day after my chemotherapy treatment, I did receive (pegfilgrastim), and that caused so much bone pain. I think I tried to take (diphenhydramine) at the time, as they said that could help. After the (pegfilgrastim) shot, once I was on my regimen, I would go in for iv. fluids because I had trouble drinking any fluids.

    I had neutropenia, but it never delayed my treatment. There was some talk about delaying a few times, but I wanted it over as soon as possible and didn't want to delay feeling better. Regarding the risk of infection, as a household, we were very careful about who we spent time with. It was very difficult to feel connected. I felt very lonely because I felt so awful.

    Communication on chemotherapy side effects:

    The (doctors) gave me a pamphlet. The things that they said verbally were that you will lose your hair and that, if you have any side effects, let us know, because there are medications that can help with them. Any questions I asked were always answered with, “Everybody responds differently, and you may have no reactions and feel great tomorrow”, so it's a case of just showing up and not knowing what to expect. I didn't want to read through all the information because there was no telling whether I would get any of the side effects, so I didn't want to put anything in my head. I went in blindly, which, looking back, probably wasn't the best thing either.

    Side effects are often dismissed by medical professionals. I think they see it so much that they forget how life-altering it is for patients. I remember bringing some things to the attention of my doctors, like the burning in my throat, and trouble eating and drinking, but they just said, “That will get better.” Those little things are so life-changing because they stop you from feeling normal, being able to engage in life, and being able to be present in the way you would like. There's no real discussion about the side effects and what can be done.

    I gained weight during chemotherapy from the steroids and because the only foods I could tolerate were carbohydrates, so I wasn't getting a balanced diet. But then, after chemotherapy, my stomach was a wreck from destroying my entire microbiome. I had chronic diarrhea for 6 months and couldn't eat due to the nausea, so then I lost a lot of weight, and it was very hard for me to put weight back on. That was never talked about.

    Financial impact of cancer:

    It was expensive: needing to pay for assistance with food and cleaning, and childcare babysitting costs that we hadn't anticipated. Cancer is expensive. When you're that sick, there was no way I could have worked. My husband took me to every treatment, there and home: I would not have been able to drive myself.

    Chemotherapy-induced myelosuppression is often managed with dose delays or reductions, but these can affect the efficacy of treatment and negatively impact patient outcomes [5,6]. Prophylactic (preventive) drugs for neutropenia are available in the form of granulocyte colony-stimulating factors (G-CSF), and reactive/therapeutic treatments include G-CSF for neutropenia, and erythropoiesis-stimulating agents (ESAs) and blood transfusions for anemia and thrombocytopenia [7–11]. Although these supportive care interventions have been shown to improve symptoms and QoL for patients receiving myelosuppressive therapy [12–14], some symptoms may not be completely alleviated. Furthermore, these treatments, specifically G-CSF, can cause bone pain and other side effects that negatively impact QoL, and ESAs carry a boxed warning [15,16].

    In 2021, the US FDA approved trilaciclib to decrease the incidence of chemotherapy-induced myelosuppression in adult patients when administered prior to an etoposide/platinum- or topotecan-containing regimen for extensive-stage small-cell lung cancer (ES-SCLC) [17]. Trilaciclib is administered intravenously to patients up to 4 h before the start of each chemotherapy treatment and works by temporarily stopping HSPCs from dividing, thus protecting them from the toxic effects of chemotherapy (myeloprotection) [17–19]. After chemotherapy treatment, when the effects of trilaciclib have worn off, HSPCs can continue to divide and develop into blood cells [18,19]. This proactive mechanism is different to other supportive care therapies, which are administered to treat myelosuppression after the damage to HSPCs has already occurred [9].

    In clinical studies, administering trilaciclib before chemotherapy resulted in reductions in the duration and occurrence of severe (grade 4) neutropenia, the rate of chemotherapy-induced anemia and thrombocytopenia, and the need for supportive care interventions [20–24]. The protective effects of trilaciclib on blood cells were reflected in improved patient experiences, as assessed using patient-reported outcome (PRO) measures such as the Functional Assessment of Cancer Therapy-Lung (FACT-L) and FACT-Anemia (FACT-An) questionnaires, which are designed to assess health-related QoL (HRQoL). Compared with patients receiving placebo, patients receiving trilaciclib reported significant improvements in several aspects of HRQoL [23].

    Several investigational agents are currently in clinical development for the management of chemotherapy-induced myelosuppression, including plinabulin, roxadustat, romiplostim, avatrombopag, eltrombopag and hetrombopag. Details of these agents are summarized in Table 1.

    Table 1. Investigational agents in development for the management of chemotherapy-induced myelosuppression.
    AgentTreatment of neutropenia/anemia/ thrombocytopeniaMechanism of actionData/registration statusRef.
    PlinabulinNeutropeniaIntravenous, marine-derived small molecule
    Blocks the synthesis of microtubules, increases numbers of HSPCs and enhances antitumor immune response by augmenting maturation of dendritic cells and activation of T cells
    Phase III study in patients with breast cancer (NCT03294577; Protective-2): reduced occurrence and duration of severe neutropenia in first week of treatment vs pegfilgrastim alone
    Breakthrough therapy designation based on interim results from Protective-2 study
    CRL issued by US FDA, requesting evidence from a further clinical study to support the NDA
    Phase III study in patients with solid tumors (NCT03102606; Protective-1): comparable efficacy, significantly less bone pain, fewer treatment delays/discontinuations and better immunosuppressive profile vs pegfilgrastim
    Phase III study in patients with advanced NSCLC (NCT02504489): improved QoL after ten docetaxel treatment cycles with plinabulin vs placebo
    [25–28]
    RoxadustatAnemiaOral hypoxia-inducible factor prolyl hydroxylase inhibitor
    Promotes erythropoiesis by increasing endogenous production of erythropoietin
    Phase III study in patients with CKD-related anemia (NCT01887600): significant increase in hemoglobin levels compared with placebo; no difference on patient QoL
    CRL issued by US FDA, requesting an additional clinical study in CKD-related anemia
    Phase II study in patients with non-myeloid malignancies with chemotherapy-induced myelosuppression (NCT04076943): increased hemoglobin levels regardless of tumor type and chemotherapy regimen
    [29–31]
    Romiplostim (NPLATE®)ThrombocytopeniaSubcutaneous Fc-peptide fusion protein
    Binds to and activates the thrombopoietin receptor, stimulating increased production of platelets
    US FDA-approved for the treatment of immune thrombocytopenia
    Phase II study in patients with solid tumors with chemotherapy-induced myelosuppression (NCT02052882): 93% of romiplostim-treated patients experienced correction of platelet count within 3 weeks vs 12.5% of control-treated patients
    Included in NCCN Guidelines® for hematopoietic growth factors as an option for the treatment of chemotherapy-induced thrombocytopenia (not US FDA approved)
    [32–35]
    Avatrombopag (DOPTELET®)ThrombocytopeniaOral thrombopoietin receptor agonist
    Binds to and activates the thrombopoietin receptor, stimulating increased platelet production
    US FDA-approved for the treatment of thrombocytopenia in adult patients with chronic liver disease who are scheduled to undergo a procedure, and for the treatment of thrombocytopenia in adults with chronic immune thrombocytopenia who have had an insufficient response to a previous treatment
    Orphan drug designation for the potential treatment of chemotherapy-induced thrombocytopenia in 2019
    Phase III study in patients with non-hematological malignancies with chemotherapy-induced thrombocytopenia (NCT03471078): similar proportions of patients not requiring platelet transfusion or chemotherapy dose reduction/delay in the avatrombopag and placebo groups
    [36–38]
    EltrombopagThrombocytopeniaOral thrombopoietin receptor agonist
    Binds to and activates the thrombopoietin receptor, stimulating increased platelet production
    Phase II study in patients with cancer receiving multiple cycles of chemotherapy (NCT00102726): similar difference in platelet count from day 1, cycle 2 to cycle 2 platelet nadir in the eltrombopag and placebo groups; greater increase in post nadir platelet counts during cycles 1 and 2 with eltrombopag compared with placebo
    Phase I/II study in patients with solid tumors receiving gemcitabine monotherapy or combination gemcitabine plus carboplatin or cisplatin (NCT01147809): shortened time to recovery from platelet nadir and fewer dose delays/reductions with eltrombopag compared with placebo
    [39]
    HetrombopagThrombocytopeniaOral thrombopoietin receptor agonist
    Binds to and activates the thrombopoietin receptor, stimulating increased platelet production
    Phase III study in patients with chemotherapy-induced thrombocytopenia receiving chemotherapy for the treatment of solid tumors (NCT03976882): ongoing[39]

    CKD: Chronic kidney disease; CRL: Complete response letter; HSPC: Hematopoietic stem and progenitor cell; NCCN: National Comprehensive Cancer Network; NDA: New drug application; NSCLC: Non-small-cell lung cancer; QoL: Quality of life.

    The aims of this review are to describe the burden of chemotherapy-induced myelosuppression on patients, and detail the strengths and limitations of current and future management strategies, with a focus on how they may improve patient QoL.

    Chemotherapy-induced neutropenia

    Neutropenia occurs when the number of neutrophils in the blood, or absolute neutrophil count (ANC), drops below normal levels (i.e., lower than 2000 cells per mm3 of blood), with severity classified according to the ANC measurement. When a person's neutrophil count falls below 1000 cells per mm3 of blood and is accompanied by a fever, this is defined as febrile neutropenia (FN) (Table 2) [40].

    Table 2. Definition, signs and symptoms and patient burden of neutropenia, anemia and thrombocytopenia.
    CytopeniaDefinitionSigns and symptomsPatient burdenRef.
    Neutropenia and FNNeutropenia:
    A finding based on laboratory test results that indicate a decrease in number of neutrophils in a blood specimen
    • Grade 1: ANC <2000–1500
    • Grade 2: ANC <1500–1000
    • Grade 3: ANC <1000–500
    • Grade 4: ANC <500

    FN (grade 3):
    A disorder characterized by an ANC <1000 and a single temperature of >38.3°C (101°F) or a sustained temperature of ≥38°C (100.4°F) for more than 1 h
    • Grade 4: Life threatening
    Fatigue
    Persistent or chronic infections
    Nausea/vomiting
    Sore throat
    Mouth sores
    Skin rashes
    Loss of appetite
    Chills or sweating
    Fever (FN)
    Chemotherapy dose reductions and treatment delays
    Increased fatigue; reduced physical, functional and emotional wellbeing
    Increased worry about risk of infection; reduced social/family wellbeing
    Hospitalization and increased risk of mortality (FN)
    Time and financial burdens associated with hospitalizations and/or additional visits to the clinic for G-CSF administration
    [40–43]
    AnemiaA disorder characterized by a reduction in the amount of hemoglobin in 100 ml of blood
    • Grade 1: Hgb <LLN-10.0
    • Grade 2: Hgb <10.0–8.0
    • Grade 3: Hgb <8.0
    • Grade 4: Hgb <6.5
    Fatigue
    Lethargy and poor concentration
    Shortness of breath
    Heart palpitations or murmurs
    Vertigo
    Loss of appetite
    Increased fatigue; reduced physical, functional and emotional wellbeing
    Chemotherapy dose reductions and treatment delays
    Financial burdens associated with RBC transfusion visits:
    • Lost wages
    • Food costs
    • Childcare expenditure
    • Transportation
    • Parking
    • Hotel accommodation
    Time burdens associated with RBC transfusion visits:
    • Chair time
    • Travel
    [40,44–46]
    ThrombocytopeniaA finding based on laboratory test results that indicate a decrease in number of platelets in a blood specimen
    • Grade 1: PLT <LLN-75,000
    • Grade 2: PLT <75,000–50,000
    • Grade 3: PLT <50,000–25,000
    • Grade 4: PLT <25,000
    Bruising
    Bleeding
    Nosebleeds/bleeding gums
    Black or bloody-looking bowel movements
    Red or pink urine
    Blood in vomit
    Heavy menstrual periods
    Severe headaches
    Muscle/joint pain
    Feeling weak or dizzy
    Chemotherapy dose reductions and treatment delays
    Risk of severe bleeding
    Anxiety and fear associated with bleeding/bruising risk and dose reductions/delays
    Increased fatigue; reduced physical, functional and emotional wellbeing
    Risk of transfusion reactions and infections with platelet transfusions
    [40,47–49]

    ANC: Absolute neutrophil count (cell/mm3); FN: Febrile neutropenia; G-CSF: Granulocyte colony-stimulating factor; Hgb: Hemoglobin (g/dl); LLN: Lower limit of normal; PLT: Platelet count (platelets/mm3); QoL: Quality of life; RBC: Red blood cell.

    Risk factors for the development of neutropenia or FN include treatment with myelosuppressive chemotherapies; advanced-stage cancer; cancers that directly affect the bone marrow, such as leukemia, lymphoma and multiple myeloma; radiation therapy targeted at areas of the body that produce bone marrow; older age (≥70 years) and comorbidities (other conditions) that impair the immune system, such as HIV or hepatitis infections [50].

    Severe neutropenia and FN often result in hospitalization and are associated with an increased risk of sepsis and death. Hospitalized patients with lung cancer experience higher rates of FN-related mortality compared with hospitalized patients with other solid tumors. Patients with lung cancer who develop FN tend to be older, are more likely to have multiple comorbidities, and have a greater incidence of pneumonia, each of which are independent risk factors for mortality, as are sepsis, infection and intensive care unit stay [51]. In addition to the risk of death, neutropenia-related hospitalizations present a major time and financial burden to patients and their caregivers, affecting their normal daily activities, and incurring costs due to travel expenses and lost work time [43].

    Lower ANC is associated with worse HRQoL [2,43,52]. In a survey of 301 patients with chemotherapy-induced myelosuppression, including 59% with neutropenia, patient-reported symptoms such as fatigue and worry about the risk of infection affected their ability to be physically active, complete work or continue meaningful relationships with friends and family [1,53]. Furthermore, in a separate study of 872 patients with advanced non-small-cell lung cancer (NSCLC), patients who developed severe neutropenia reported significantly increased fatigue and nausea/vomiting, significantly worse physical, social and cognitive functioning, and increased arm and shoulder pain compared with those without severe neutropenia [54].

    In addition to generalized QoL measures such as the FACT-General (FACT-G) questionnaire for measuring physical, social, emotional and functional wellbeing, several neutropenia-specific tools have been developed, which may provide a better understanding of QoL in patients with chemotherapy-induced neutropenia. The most notable is FACT-Neutropenia (FACT-N), a 19-item questionnaire used in addition to the FACT-G, which focuses on neutropenia-specific issues such as malaise, worry and flu-like symptoms [55,56].

    The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Hematopoietic Growth Factors (Version 1.2024) recommend the prophylactic use of G-CSF (filgrastim, pegfilgrastim or biosimilars) for the management of chemotherapy-induced neutropenia, as well as the therapeutic use of G-CSF in patients who have already developed FN [35]. For patients with ES-SCLC, NCCN Guidelines® include trilaciclib as a prophylactic option to decrease the incidence of chemotherapy-induced myelosuppression when administered before platinum/etoposide ± immune checkpoint inhibitor-containing regimens or a topotecan-containing regimen [35].

    G-CSF is a type of medication often used to stimulate the production of WBCs, including neutrophils, which are important in fighting infection [57]. For prophylactic management with G-CSF, a patient's risk for developing FN is assessed on the basis of type and stage of cancer, chemotherapy regimen, patient risk factors and whether the treatment is curative or palliative. If a patient has a high risk of FN (>20%), prophylactic use of G-CSF after the first chemotherapy cycle is recommended. For patients with an intermediate FN risk (10–20%), prophylactic G-CSF use is considered on the basis of risk factors such as older age, prior chemotherapy or radiation therapy, persistent neutropenia and bone marrow involvement by the tumor [7,8]. The risk of FN in the first chemotherapy cycle is reported to be greater in patients who receive pegfilgrastim on the same day as chemotherapy than in those who receive it 1–4 days post chemotherapy [41] and, in the US, G-CSF is typically given during separate visits to those for myelosuppressive chemotherapy [42].

    Therapeutic use of filgrastim (or filgrastim biosimilars) is recommended for patients already presenting with FN who have previously received these treatments in a preventive manner; however, the therapeutic use of G-CSF is not recommended in those who have previously received long-acting G-CSF (pegfilgrastim or pegfilgrastim biosimilars). For patients with FN who have not received any prophylactic G-CSF, assessment of risk factors associated with infection-related complications or poor clinical outcomes is recommended [7,8].

    Although data are scarce on the impact of G-CSF use on QoL, patients who receive G-CSF prophylaxis are likely to have better HRQoL owing to: their ability to maintain their relative dose of chemotherapy; a reduced need for antibiotic treatment; a reduced likelihood of hospitalization or death due to FN; and a reduced impact on aspects of daily life and work [15,58,59].

    However, there are also side effects associated with G-CSF that can negatively affect patients' QoL. The most common side effect of G-CSF is bone pain, which occurs mainly because the medication stimulates the production of neutrophils, causing pain or discomfort in the bones [15,60]. The severity of bone pain can vary from person to person and may be most notable in the bones of the spine, pelvis and legs [61]. In addition, bone pain may be accompanied by other symptoms, such as fatigue, fever or muscle aches [15,57,61]. Bone pain was reported by our patient author as a severe side effect of treatment with pegfilgrastim (Box 1). In most cases, bone pain can be managed by other medications [15,60]. However, patients should report any persistent or severe pain to a healthcare provider [62]. In addition to interfering with daily activities, bone pain can lead to dose delays or discontinuation of treatment [15], which can have a negative impact on patients and their friends and family. Pegfilgrastim is approved for next-day use, and, unless the on-body injector is used, additional appointments further complicate the lives of the patient and caregiver.

    In addition to current US FDA approvals for G-CSF and trilaciclib, other agents that may impact myelosuppression are under development. In September 2022, the US FDA-approved eflapegfilgrastim-xnst, a long-acting G-CSF with a novel formulation to decrease the incidence of infection, as manifested by FN, in adult patients with non-myeloid malignancies receiving myelosuppressive anticancer drugs associated with clinically significant incidence of FN [63,64].

    Plinabulin is an investigational small molecule in development for the management of chemotherapy-induced neutropenia (Table 1). In addition to showing efficacy in phase III trials, plinabulin was associated with significantly less patient-reported bone pain compared with pegfilgrastim (difference, -0.67; 95% CI, -1.17 to -0.16; p = 0.01), as well as resulting in fewer chemotherapy dose delays of >7 days (3.8 vs 5.7%) and discontinuations (13.5 vs 26.4%), and a better immunosuppressive profile (neutrophil-to-lymphocyte ratio of >5 at day 8, 3.8 vs 46.0%; p < 0.001) [25,27]. Additionally, plinabulin was shown to improve QoL in a phase III study in patients with advanced NSCLC treated with docetaxel, in which QoL was assessed using the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-Cancer Core 30-item (QLQ-C30) and Lung Cancer 13-item (QLQ-LC13) measures. Outcomes were comparable between plinabulin and placebo during the first ten chemotherapy cycles; however, after the first ten cycles, patients who received plinabulin had more favorable cumulative EORTC QLQ-C30 and QLQ-LC13 scores, and significant improvements in symptoms, including mouth sores, dysphagia and coughing [28].

    ALRN-6924 (sulanemadlin) is an investigational molecule that increases the activity of the p53 tumor suppressor protein. The role of p53 is to stop cells from dividing too fast and becoming cancerous. p53 is present in healthy cells, such as HSPCs in the bone marrow. Treatment with ALRN-6924 stops HSPCs from dividing, potentially protecting them from the toxic effects of chemotherapy [65,66]. However, in a phase Ib study (NCT05622058), patients receiving ALRN-6924 plus chemotherapy for the treatment of p53-mutated breast cancer experienced grade 4 neutropenia and alopecia. The study failed to meet its primary and secondary end points of duration and incidence of severe neutropenia in cycle 1 and incidence of chemotherapy-induced alopecia, respectively, and the study was terminated [67]. Moreover, further development of ALRN-6924 was stopped, including the termination of clinical trials in p53-mutated ES-SCLC and NSCLC [67].

    G-CSF stimulates stem cell growth and neutrophil production after the bone marrow has received chemotherapy, raising concerns about a risk of longer-term damage, including conditions such as myelodysplasia and leukemia, which are known risks of chemotherapy [68]. By contrast, trilaciclib has been shown to proactively protect the bone marrow from chemotherapy-induced damage, resulting in longer-term protection of HSPCs [19]. Pooled data from the three clinical studies evaluating trilaciclib in patients with ES-SCLC showed that administering trilaciclib before chemotherapy significantly reduced the duration and occurrence of severe neutropenia compared with placebo, regardless of whether patients had received G-CSF [24]. Although the frequency of FN was low across the studies, there was a threefold decrease in its occurrence among patients who received trilaciclib. This translated into improvements in patients' QoL, with patients who received trilaciclib reporting significant improvements in physical and functional wellbeing, and in the symptoms and impact of fatigue [23].

    Overall, therefore, by expanding upon existing treatment options, the inclusion of trilaciclib in treatment guidelines and the development of the investigational agent plinabulin are important advances in supportive care for chemotherapy-induced neutropenia. Further studies of these agents in other settings are eagerly awaited.

    Chemotherapy-induced anemia

    The circulatory system is responsible for distributing oxygen throughout the body. This is achieved by RBCs, which contain the oxygen-binding molecule hemoglobin. A normal level of hemoglobin is 12–16 g per 100 ml (g/dl) of blood for women and 14–18 g/dl for men, and anemia is defined by a reduction in the amount of hemoglobin in the blood below the lower limit of normal (Table 2) [40,44].

    A drop in hemoglobin count may be caused by a reduced ability of the body to make new RBCs (e.g., because of nutritional deficiencies), loss of RBCs through bleeding, or destruction of RBCs. People with cancer have a greater risk of anemia compared with healthy individuals, even before starting cytotoxic chemotherapy, and having advanced-stage cancer or nutritional deficiencies associated with cancer (such as iron, folate or B12 deficiencies) are risk factors for developing this condition [69].

    Chemotherapy-induced anemia is a common side effect of platinum-based chemotherapy treatments such as cisplatin, carboplatin or oxaliplatin. The production of RBCs in the bone marrow (erythropoiesis) is usually stimulated by a chemical called erythropoietin, which is produced in the kidneys. Platinum-based drugs can damage the erythropoietin-producing cells in the kidneys and directly suppress HSPCs in the bone marrow, resulting in reduced RBC production. Among patients with solid tumors, chemotherapy-induced anemia is particularly common in patients with lung cancer, as they are often treated with platinum-based chemotherapy, tend to be older and frequently have pulmonary or cardiovascular comorbidities that can make anemia symptoms worse [44,70].

    Symptoms of chemotherapy-induced anemia include lethargy and poor concentration, shortness of breath (dyspnea), heart palpitations or murmurs, vertigo and loss of appetite. However, the most common symptom (in 60–90% of patients) is fatigue, which can have major adverse effects on patients' QoL [44,45], as reflected in the experiences of our patient author (Box 1). When patients with cancer are asked to rank which side effects of chemotherapy have the greatest impact on their lives, fatigue is often reported as being the most debilitating across all cancer types [45,71,72]. Participants of the 2010 LIVESTRONG Survey, which included 3129 cancer survivors, reported ‘energy and rest’ as being the most common physical concern and causing the most functional impairment. However, less than 20% of respondents who reported having concerns with energy indicated that they had received care to address this concern [72]. Another survey, of 152 patients with various solid tumors, found that patients with anemia reported significantly worse physical, social and role functioning, and experienced more fatigue, sickness and vomiting and loss of appetite compared with patients without anemia. In addition, low hemoglobin levels were associated with worse global QoL and fatigue, and higher depression scores [73].

    The impact of anemia and fatigue on patients can be assessed using PRO measures such as the FACT-An or Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) questionnaires [74]. FACIT-F includes the 27-item FACT-G and a 13-item questionnaire designed to assess the effects of fatigue in patients with cancer. FACT-An includes the 40-item FACIT-F plus an additional seven items related to anemia [48]. Several studies have shown that increases in patients' hemoglobin levels correlate with improvements in FACT-G, FACIT-F and FACT-An scores, reflecting improved QoL [14,75–77]. In an analysis of five clinical studies of patients with cancer who had anemia, patients whose hemoglobin levels had increased reported significantly greater improvements in FACIT-F subscale scores, which translated into greater energy and activity levels and improved self-perception of overall health [76]. Similarly, results from a phase IV study of patients with solid tumors who had received treatment for chemotherapy-induced anemia (NCT01444456; eAQUA) showed that an increase in hemoglobin levels by >1 g/dl was associated with improved QoL, as measured by changes in FACT-F scores [14]. More recently, a study of 133 chemotherapy-treated patients with cancer found that hemoglobin levels correlated with self-reported QoL scores for fatigue, physical and functional wellbeing and performance status [13].

    Current NCCN Guidelines® recommend using RBC transfusions, ESAs or iron supplementation for the management of chemotherapy-induced anemia. In addition, trilaciclib is a prophylactic option to decrease the incidence of anemia and RBC transfusions when administered before platinum/etoposide with or without immune checkpoint inhibitor-containing regimens or a topotecan-containing regimen for ES-SCLC [35]. In contrast to European guidelines, the choice of treatment is not based on a certain hemoglobin threshold, and RBC transfusions are reserved mainly for high-risk and/or symptomatic patients. ESAs are recommended on the basis of patient preference, with the goals of gradual symptom improvement and avoidance of transfusion, whereas iron supplementation is recommended for absolute or functional iron deficiency [9,35].

    RBC transfusions can result in an immediate increase in hemoglobin levels, providing rapid relief from the symptoms of chemotherapy-induced anemia [9]. However, transfusions carry the risk of transfusion-related reactions, volume overload or iron overload, and have the added burden of time spent in the clinic and the associated travel [9,46]. Results from a study of 103 patients with cancer found that the average chair time for patients receiving RBC transfusions was 4 h, and the average time spent traveling to and from the clinic was approximately 1 h. In addition to chair and travel time, RBC transfusions were associated with indirect financial costs to patients, such as lost wages, food costs, childcare expenditures, hotel accommodation and transportation or parking costs [46]. Furthermore, the hemoglobin threshold level at which RBC transfusions are considered has become lower over time (<7.0–8.0 g/dl compared with a historic threshold of <9.0–10.0 g/dl), which negatively impacts the QoL of patients who are receiving myelosuppressive chemotherapy agents for cancer treatment.

    In a systematic review of studies measuring HRQoL in patients with chemotherapy-induced anemia, 34 of 35 studies reported significant improvements in patient QoL following ESA treatment, as assessed by FACT-F, FACT-An, Linear Analog Scale Assessment, and QLQ-C30, compared with patients who did not receive ESAs [78]. Among 114 patients treated with the ESA epoetin beta during cancer treatment, increased hemoglobin levels were associated with improved HRQoL, measured using visual analogue scale scores for fatigue [79]. Furthermore, in a study of 223 patients receiving myelotoxic chemotherapy for breast cancer, patients who received epoetin alfa had greater improvements in hemoglobin levels and significant improvements in FACT-An anemia and fatigue subscales compared with those who received best standard care [80]. In a separate study of 269 patients with chemotherapy-related anemia, early treatment of mild anemia with epoetin alfa significantly improved hemoglobin levels, QoL and productivity compared with delaying treatment until hemoglobin level decreased to <9.0 g/dl [81]. Finally, treatment with darbepoetin alfa has been associated with an improvement in symptom burden as measured by the MD Anderson Symptom Inventory [82].

    ESA therapy reduces the need for transfusions and can relieve symptoms; however, unlike RBC transfusions, it can take several weeks for ESA therapy to stimulate an increase in hemoglobin levels. There are additional risks associated with ESAs, which can make physicians reluctant to use them, including an increased risk of thromboembolic events, hypertension and potential increased mortality and tumor progression [9,10]. In 2007, the US FDA approved a boxed warning for ESA products and, in 2010, introduced the Risk Evaluation and Mitigation Strategy (REMS) implementation for ESA use. This required medical professionals to be REMS-certified to be able to use ESAs and required patients to complete a consent form describing the benefits and risks of ESAs. The REMS program was discontinued in 2017, as it was no longer considered necessary for ensuring that the benefits of ESA therapy outweigh the risks in patients with cancer [83]. However, the US FDA label carries a boxed warning stating that, in patients with cancer, ESAs should be used only for the treatment of chemotherapy-induced anemia, be discontinued upon completion of the chemotherapy course, and not be used when treatment is curative in intent [16]. Prescription of ESAs fell significantly in the USA when safety concerns were first reported, and there is evidence to show a further reduction in ESA use after changes in reimbursement were brought into effect in 2008 [84]. However, there is also evidence that the implementation of the REMS program had little effect on ESA administration [83,85].

    Both RBC transfusions and ESAs are administered after anemia is already apparent, whereas trilaciclib can provide prophylactic protection from chemotherapy-induced anemia. In a pooled phase III analysis, administering trilaciclib prior to chemotherapy significantly reduced the occurrence of grade 3 or 4 anemia, the use of ESAs, and the need for RBC transfusions on or after week 5 of study treatment [23,24]. The frequency of RBC transfusions was approximately halved with the administration of trilaciclib, which may translate into reduced patient burden in terms of the additional time and costs associated with transfusion visits. Additionally, patients receiving trilaciclib reported significant improvements in symptoms associated with anemia, as assessed by FACT-An [23].

    Roxadustat is an orally active hypoxia-inducible factor prolyl hydroxylase inhibitor that was investigated for the treatment of chronic kidney disease-related anemia in patients not on dialysis (Table 1). Results from a phase III trial in this patient population showed that administering roxadustat significantly increased hemoglobin levels compared with placebo, although there were no differences in PROs between the two treatment groups [29]. Roxadustat has also been shown to increase hemoglobin levels in patients with non-myeloid cancers and chemotherapy-induced anemia, regardless of the tumor type and chemotherapy regimen. In this study, fewer patients required an RBC transfusion on or after week 5 when starting on 2.5 mg/kg compared with a lower 2.0 mg/kg dose [31].

    Chemotherapy-induced thrombocytopenia

    Platelets, also known as thrombocytes, are a type of blood cell that help the blood to clot (thrombosis). Thrombocytopenia is defined by a lower-than-normal number of platelets per mm3 of blood (Table 2). The incidence of thrombocytopenia varies according to the type of chemotherapy, being more common in patients receiving highly myelosuppressive treatments such as gemcitabine- and platinum-based chemotherapies [39,86]. Chemotherapy drugs can cause thrombocytopenia in different ways, with some reducing platelet production through effects on HSPCs and others increasing the rate of platelet destruction [39,87].

    There is a shortage of literature describing the impact of chemotherapy-related thrombocytopenia on patients' QoL. However, having a low platelet count can potentially cause serious problems for patients with cancer, as it increases the risk of bleeding. At platelet counts lower than 10,000 per microliter (μl) of blood, the risk of spontaneous bleeding is increased, and surgical procedures are often complicated by bleeding at platelet counts <50,000/μl. Even at platelet counts <100,000/μl, chemotherapy and radiation therapy should be administered carefully to avoid making the thrombocytopenia worse and increasing the bleeding risk [87]. In addition to bleeding, other symptoms of a low platelet count include bruising, small purple or red dots under the skin (called petechiae), severe headaches and blood in the urine, vomit or bowel movements [47]. Having a diagnosis of thrombocytopenia has been reported to worsen patients' feelings of anxiety and fear beyond those associated with the cancer diagnosis [39,87]. Moreover, the costs of care related to chemotherapy-induced thrombocytopenia can be substantial [86], creating an additional burden on patients, caregivers and healthcare systems.

    PROs used to measure the impact of thrombocytopenia on patients' QoL include the FACT-Thrombocytopenia 18-item version (FACT-Th18), which is composed of the FACT-G and an 18-item thrombocytopenia subscale that includes questions relating to bleeding, bruising, activity and concerns about bleeding or dose reductions/delays. Two shorter versions with reduced thrombocytopenia subscales are also available in the form of the FACT-Th11 (11 items) and FACT-Th6 (6 items) [48,88].

    Chemotherapy-induced thrombocytopenia is commonly managed with treatment delays or dose reductions, which aim to allow bone marrow recovery [49]. Changes to planned treatment can cause inconvenience and/or additional worry to patients, as reflected in the FACT-Th questionnaires [48]. Platelet transfusions are an effective and rapid treatment for severe thrombocytopenia and are recommended in AABB (Association for the Advancement of Blood & Biotherapies; formerly, the American Association of Blood Banks) guidelines to reduce the risk of spontaneous bleeding in patients with therapy-induced thrombocytopenia [49,89]. However, platelet transfusions offer only a temporary improvement in platelet counts [34]. Additionally, transfusions may be ineffective in some patients and/or cause febrile or allergic reactions. Platelet transfusions are also associated with a risk of infection due to bacterial contamination [49,90].

    Although not FDA-approved for chemotherapy-induced thrombocytopenia, NCCN Guidelines® for Hematopoietic Growth Factors also recommend romiplostim, or enrollment in a clinical trial of another thrombopoietin receptor agonist such as avatrombopag, eltrombopag or hetrombopag, as alternative options to maintain the dose schedule and intensity of chemotherapy if the benefit is expected to outweigh the risks [35,39]. Romiplostim is an Fc-peptide fusion protein that binds to and activates the thrombopoietin receptor, stimulating increased platelet production (Table 1). It is FDA approved and widely used to treat immune thrombocytopenia [32,33]. Additionally, romiplostim has shown efficacy in improving platelet counts among patients with solid tumors and chemotherapy-induced myelosuppression in a phase II randomized trial, as well as several retrospective studies and case series [32,34]. In the phase II study, which included patients who had platelet counts of <100,000/μl for at least 4 weeks, treatment with romiplostim for 2 weeks (n = 52) resulted in a mean platelet count of 141,000/μl compared with 57,000/μl after 3 weeks in the untreated observation group (n = 8) [32]. In one retrospective study that included 153 patients with solid tumors, romiplostim allowed 79% of patients to avoid further chemotherapy dose reductions/treatment delays and 89% avoided platelet transfusions [34]. It should be noted, however, that thrombopoietin receptor agonists are associated with an increased risk of venous thromboembolism and should be used with caution in patients with cancer [34]. To our knowledge, the impact of romiplostim on QoL has not been investigated in patients with chemotherapy-induced thrombocytopenia, although patients with chronic immune thrombocytopenia have reported better HRQoL following romiplostim therapy in phase III trials, including improvements in measures of symptoms, fear and activity [91].

    Conclusion

    Not only can myelosuppressive side effects affect the ability to deliver planned chemotherapy treatment, potentially adversely affecting clinical outcomes, but they can also have a substantial negative impact on patients' HRQoL. Despite the availability of G-CSF, ESAs and transfusions, chemotherapy-induced myelosuppression is associated with a significant QoL burden on patients and their caregivers, with symptoms resulting in impairment of normal daily functioning and worries over infection or bleeding risk. Traditional supportive care measures are also often used reactively, after symptoms have already emerged, and are each associated with certain risks and limitations that can further exacerbate the QoL burden.

    Administering trilaciclib prior to chemotherapy has been shown to result in clinically meaningful reductions in myelosuppression and its symptoms, as well as fewer hospitalizations and a reduced need for supportive care interventions. These effects translate into improvements in HRQoL measures related to the protected cell lineages, including fatigue and physical and functional wellbeing [23]. Several pipeline drugs such as plinabulin and roxadustat have also shown encouraging results in clinical trials and have the potential to improve QoL for patients on chemotherapy, both by improving blood counts to directly alleviate symptoms, and indirectly by reducing the need for G-CSF, ESAs, or transfusions.

    Future perspective

    Following on from the approval of trilaciclib in patients with ES-SCLC in 2021, the myeloprotective benefit of trilaciclib is being investigated in clinical trials in other cancer types, including advanced bladder cancer and breast cancer. Additionally, it is likely that several pipeline agents, some of which are already recognized by the FDA as having potential utility for managing chemotherapy-induced myelosuppression, will also be commercially available for the prevention/treatment of one or more cytopenias. The availability of more treatment options in the future will likely expand our current supportive care of G-CSF, ESAs, trilaciclib and transfusions, to include new indications and options, ultimately improving outcomes for our patients.

    Executive summary
    • Chemotherapy-induced damage of hematopoietic stem and progenitor cells in the bone marrow can result in myelosuppression (neutropenia, anemia and/or thrombocytopenia), which adversely affects patients' quality of life.

    • Chemotherapy-induced myelosuppression is commonly managed with chemotherapy dose reductions or delays, which can negatively impact treatment outcomes, or supportive care interventions that are each associated with risks and limitations.

    Chemotherapy-induced neutropenia

    • Neutropenia symptoms such as fatigue and concerns over the risk of infection negatively affect patients' daily activities and relationships.

    • Granulocyte colony-stimulating factors reduce the risk of severe or febrile neutropenia but commonly lead to bone pain, which may result in chemotherapy dose delays or discontinuation.

    Chemotherapy-induced anemia

    • Lower hemoglobin levels among patients with chemotherapy-induced anemia are correlated with poorer quality of life, and the most common symptom, fatigue, can be extremely debilitating for patients.

    • Erythropoiesis-stimulating agents are associated with thromboembolic events, hypertension and potential increased mortality and tumor progression, while red blood cell transfusions carry the risk of transfusion-related reactions, volume overload or iron overload.

    Chemotherapy-induced thrombocytopenia

    • Symptoms of a low platelet count include bruising and bleeding, and a diagnosis of thrombocytopenia can worsen patients' feelings of anxiety and fear beyond those associated with the cancer diagnosis.

    • Platelet transfusions may be used in severe cases of thrombocytopenia but offer only a temporary solution and may cause infections, and febrile or allergic reactions.

    Conclusions & future perspectives

    • Unlike conventional supportive care options, trilaciclib is used proactively to reduce myelosuppression and its symptoms across multiple blood cell lineages, which translates into improvements in patients' quality of life.

    • In addition, several pipeline drugs have shown encouraging results in clinical trials and have the potential to improve quality of life for patients on chemotherapy.

    Author contributions

    All authors contributed to the conception or design of the work; or the acquisition, analysis, or interpretation of data for the work. All authors contributed to drafting the work or revising it critically for important intellectual content. All authors provided final approval of the version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

    Financial disclosure

    This work was funded by G1 Therapeutics, Inc. J Crawford has received institutional research funding from AstraZeneca, Helsinn, the National Cancer Institute's National Clinical Trials Network, and Pfizer. J Weiss has received consulting fees and research funding from G1 Therapeutics, Inc. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    J Crawford has participated as a scientific advisor for Actimed Therapeutics, Enzychem Lifesciences, Faraday Pharmaceuticals, Jazz Pharmaceuticals, Partner Therapeutics, and Pfizer; participated on publications committees for Amgen, Frensenius Kabi, G1 Therapeutics Inc., LUNGevity Foundation, Pfizer and Spectrum Pharmaceuticals; and participated on data safety monitoring boards for BioAtla, G1 Therapeutics, Inc and NiA Pharmaceuticals. The authors have no other competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Writing disclosure

    Medical writing assistance was provided by Farhana Burnett, of Envision Pharma Group, funded by G1 Therapeutics, Inc.

    Informed consent disclosure

    The authors state that they have obtained verbal and written informed consent from the patient author for the inclusion of their medical and treatment history within this review.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Epstein RS, Aapro MS, Basu Roy UK et al. Patient burden and real-world management of chemotherapy-induced myelosuppression: results from an online survey of patients with solid tumors. Adv Ther. 2020;37(8):3606–3618. doi: 10.1007/s12325-020-01419-6 •• An online survey of 301 participants with breast, lung, or colorectal cancer describing the real-world impact of chemotherapy-induced myelosuppression. Participants reported that myelosuppression had a negative impact on their daily lives and that symptoms including fatigue, weakened immune system, bleeding and/or bruising and shortness of breath were as bothersome as other side chemotherapy effects (e.g., alopecia, neuropathy and nausea/vomiting).
    • 2. Epstein RS, Weerasinghe RK, Parrish AS et al. Real-world burden of chemotherapy-induced myelosuppression in patients with small cell lung cancer: a retrospective analysis of electronic medical data from community cancer care providers. J. Med. Econ. 2022;25(1):108–118. doi: 10.1080/13696998.2021.2020570
    • 3. Barreto JN, McCullough KB, Ice LL et al. Antineoplastic agents and the associated myelosuppressive effects: a review. J. Pharm. Pract. 2014;27(5):440–446. doi: 10.1177/0897190014546108 • A comprehensive review of the mechanisms and consequences of myelosuppression, including the structure and function of normal bone marrow, process of hematopoiesis and myelotoxicities associated with common antineoplastic agents.
    • 4. Smith RE. Trends in recommendations for myelosuppressive chemotherapy for the treatment of solid tumors. J. Natl Compr. Canc. Netw. 2006;4(7):649–658. doi: 10.6004/jnccn.2006.0056
    • 5. Crawford J, Denduluri N, Patt D et al. Relative dose intensity of first-line chemotherapy and overall survival in patients with advanced non-small-cell lung cancer. Support. Care Cancer 2020;28(2):925–932. doi: 10.1007/s00520-019-04875-1
    • 6. 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. doi: 10.1002/cam4.200
    • 7. 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. doi: 10.1200/JCO.2015.62.3488
    • 8. Klastersky J, de Naurois J, Rolston K et al. Management of febrile neutropaenia: ESMO clinical practice guidelines. Ann. Oncol. 2016;27(Suppl. 5):v111–v118. doi: 10.1093/annonc/mdw325
    • 9. Aapro M, Beguin Y, Bokemeyer C et al. Management of anaemia and iron deficiency in patients with cancer: ESMO clinical practice guidelines. Ann. Oncol. 2018;29(Suppl. 4):iv271. doi: 10.1093/annonc/mdy323
    • 10. Bohlius J, Bohlke K, Castelli R et al. Management of cancer-associated anemia with erythropoiesis-stimulating agents: ASCO/ASH clinical practice guideline update. J. Clin. Oncol. 2019;37(15):1336–1351. doi: 10.1200/JCO.18.02142
    • 11. Schiffer CA, Bohlke K, Delaney M et al. Platelet transfusion for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 2018;36(3):283–299. doi: 10.1200/JCO.2017.76.1734
    • 12. Arantes LH Jr, Crawford J, Gascon P et al. A quick scoping review of efficacy, safety, economic, and health-related quality-of-life outcomes of short- and long-acting erythropoiesis-stimulating agents in the treatment of chemotherapy-induced anemia and chronic kidney disease anemia. Crit. Rev. Oncol. Hematol. 2018;129:79–90. doi: 10.1016/j.critrevonc.2018.06.010
    • 13. Yakymenko D, Frandsen KB, Christensen IJ et al. Randomised feasibility study of a more liberal haemoglobin trigger for red blood cell transfusion compared to standard practice in anaemic cancer patients treated with chemotherapy. Transfus. Med. 2018;28(3):208–215. doi: 10.1111/tme.12439
    • 14. Mouysset JL, Freier B, van den Bosch J et al. Hemoglobin levels and quality of life in patients with symptomatic chemotherapy-induced anemia: the eAQUA study. Cancer Manag. Res. 2016;8:1–10. doi: 10.2147/CMAR.S88110
    • 15. Lapidari P, Vaz-Luis I, Di Meglio A. Side effects of using granulocyte-colony stimulating factors as prophylaxis of febrile neutropenia in cancer patients: a systematic review. Crit. Rev. Oncol. Hematol. 2021;157:103193. doi: 10.1016/j.critrevonc.2020.103193
    • 16. United States Food and Drug Administration. EPOGEN® (epoetin alfa) injection, for intravenous or subcutaneous use [prescribing information]. Thousand Oaks (CA): Amgen, Inc.; July 2018 [cited 2024 Feb 16]. Available from: www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Epogen/epogen_pi_hcp_english.pdf
    • 17. United States Food and Drug Administration. COSELA® (trilaciclib) for injection, for intravenous use [prescribing information]. Durham (NC): G1 Therapeutics, Inc.; August 2023 [cited 2024 Feb 16]. Available from: www.g1therapeutics.com/file.cfm/69/docs/cosela_approved_label_8-3-2023.pdf
    • 18. Bisi JE, Sorrentino JA, Roberts PJ et al. Preclinical characterization of G1T28: a novel CDK4/6 inhibitor for reduction of chemotherapy-induced myelosuppression. Mol. Cancer Ther. 2016;15(5):783–793. doi: 10.1158/1535-7163.MCT-15-0775
    • 19. 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):eaal3986. doi: 10.1126/scitranslmed.aal3986
    • 20. 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 2021;148(10):2557–2570. doi: 10.1002/ijc.33453 •• In this pivotal randomized phase II trial, administering trilaciclib before standard-of-care chemoimmunotherapy (etoposide/carboplatin plus atezolizumab) clinically and significantly reduced the duration and occurrence of severe neutropenia and resulted in lower rates of grade 3/4 hematologic adverse events and improved health-related quality of life for patients with extensive-stage small-cell lung cancer.
    • 21. 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. doi: 10.1007/s12325-020-01538-0
    • 22. 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. doi: 10.1093/annonc/mdz278
    • 23. Weiss J, Goldschmidt J, Andric Z et al. Effects of trilaciclib on chemotherapy-induced myelosuppression and patient-reported outcomes in patients with extensive-stage small cell lung cancer: pooled results from three Phase II randomized, double-blind, placebo-controlled studies. Clin. Lung Cancer 2021;22(5):449–460. doi: 10.1016/j.cllc.2021.03.010 • This pooled analysis of three randomized phase II trials of trilaciclib in patients with extensive-stage small cell lung cancer found that administering trilaciclib before chemotherapy reduced most measures of multilineage myelosuppression, which translated into a reduced need for supportive care interventions and improvements in health-related quality of life, including measures of fatigue, physical wellbeing and functional wellbeing.
    • 24. Ferrarotto R, Anderson I, Medgyasszay B et al. Trilaciclib prior to chemotherapy reduces the usage of supportive care interventions for chemotherapy-induced myelosuppression in patients with small cell lung cancer: pooled analysis of three randomized Phase II trials. Cancer Med. 2021;10(17):5748–5756. doi: 10.1002/cam4.4089
    • 25. Blayney DW, Shi Y, Adamchuk H et al. Clinical trial testing superiority of combination plinabulin (Plin) and pegfilgrastim (Peg) versus peg alone in breast cancer treated with high-risk febrile neutropenia risk chemotherapy (chemo): final results of the Phase III protective-2 in chemo-induced neutropenia (CIN) prevention. J. Clin. Oncol. 2021;39(Suppl. 15):533. doi: 10.1200/JCO.2021.39.15_suppl.533
    • 26. BeyondSpring Pharmaceuticals, Inc. BeyondSpring receives breakthrough therapy designations from both US FDA and China NMPA for plinabulin in chemotherapy-induced neutropenia indication Florham Park (NJ): BeyondSpring Pharmaceuticals, Inc., [updated 2020 Sep 8; cited 2024 Feb 16]. Available from: https://beyondspringpharma.com/beyondspring-receives-breakthrough-therapy-designations-from-both-u-s-fda-and-china-nmpa-for-plinabulin-in-chemotherapy-induced-neutropenia-indication/
    • 27. Blayney DW, Mohanlal R, Adamchuk H et al. Efficacy of plinabulin vs pegfilgrastim for prevention of docetaxel-induced neutropenia in patients with solid tumors: a randomized clinical trial. JAMA Netw. Open 2022;5(1):e2145446. doi: 10.1001/jamanetworkopen.2021.45446
    • 28. Feinstein T, Ogenstad S, Mitchell D et al. DUBLIN-3 results on quality of life (QoL) in second/third-line EGFR-wild type NSCLC patients (pts) receiving docetaxel (Doc) with or without plinabulin (Plin) using the validated EORTC QLQ C30 and QLQ LC13 questionnaires. J. Clin. Oncol. 2022;40(Suppl. 16):9091. doi: 10.1200/JCO.2022.40.16_suppl.9091 • In this phase III study comparing plinabulin versus placebo in patients with advanced non-small-cell lung cancer (NSCLC) treated with docetaxel, outcomes were comparable during the first 10 chemotherapy cycles; however, after the first 10 cycles, patients who received plinabulin had more favorable cumulative reported statistically significant benefits in quality-of-life measures (EORTC QLQ C30 and LC13), along with improvements in symptoms, including mouth sores, dysphagia and coughing.
    • 29. Shutov E, Sułowicz W, Esposito C et al. Roxadustat for the treatment of anemia in chronic kidney disease patients not on dialysis: a Phase III, randomized, double-blind, placebo-controlled study (ALPS). Nephrol. Dial. Transplant. 2021;36(9):1629–1639. doi: 10.1093/ndt/gfab057
    • 30. FibroGen, Inc. FibroGen receives complete response letter from the FDA for roxadustat for anemia of chronic kidney disease San Francisco (CA): FibroGen, Inc., [updated 2021 Aug 11; cited 2024 Feb 16]. Available from: https://investor.fibrogen.com/news-releases/news-release-details/fibrogen-receives-complete-response-letter-fda-roxadustat-anemia
    • 31. Glaspy J, Gabrail NY, Locantore-Ford P et al. Open-label, Phase II study of roxadustat for the treatment of anemia in patients receiving chemotherapy for non-myeloid malignancies. Am. J. Hematol. 2023;98(5):703–711. doi: 10.1002/ajh.26865
    • 32. Soff GA, Miao Y, Bendheim G et al. Romiplostim treatment of chemotherapy-induced thrombocytopenia. J. Clin. Oncol. 2019;37(31):2892–2898. doi: 10.1200/JCO.18.01931
    • 33. United States Food and Drug Administration. NPLATE® (romiplostim) for injection, for subcutaneous use [prescribing information]. Thousand Oaks (CA): Amgen, Inc.; February 2022 [cited 2024 Feb 16]. Available from: www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Nplate/nplate_pi_hcp_english.pdf
    • 34. Al-Samkari H, Parnes AD, Goodarzi K et al. A multicenter study of romiplostim for chemotherapy-induced thrombocytopenia in solid tumors and hematologic malignancies. Haematologica 2021;106(4):1148–1157. doi: 10.3324/haematol.2020.251900
    • 35. National Comprehensive Cancer Network. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Hematopoietic Growth Factors Version 3.2024. © National Comprehensive Cancer Network, Inc. 2024. All rights reserved. Accessed February 16, 2024. To view the most recent and complete version of the guideline, go online to NCCN.org. NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way. •• The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Hematopoietic Growth Factors provide guidance on the evaluation, prevention and management of febrile neutropenia, chemotherapy-induced thrombocytopenia and chemotherapy-induced anemia in adult patients with cancer, especially those receiving chemotherapy.
    • 36. United States Food and Drug Administration. DOPTELET® (avatrombopag) tablets, for oral use [prescribing information]. Durham (NC): AkaRx, Inc.; July 2021 [cited 2024 Feb 16]. Available from: https://doptelet.com/themes/pdf/prescribing-information.pdf
    • 37. Sobi. FDA grants avatrombopag orphan drug designation for the treatment of chemotherapy-induced thrombocytopenia. Stockholm, Sweden: Swedish Orphan Biovitrum AB. [updated 2019 Dec 21; cited 2024 Feb 16]. Available from: www.sobi.com/en/press-releases/fda-grants-avatrombopag-orphan-drug-designation-treatment-chemotherapy-induced
    • 38. Al-Samkari H, Kolb-Sielecki J, Safina SZ et al. Avatrombopag for chemotherapy-induced thrombocytopenia in patients with non-haematological malignancies: an international, randomised, double-blind, placebo-controlled, Phase III trial. Lancet Haematol. 2022;9(3):e179–e189. doi: 10.1016/S2352-3026(22)00001-1
    • 39. Kuter DJ. Treatment of chemotherapy-induced thrombocytopenia in patients with non-hematologic malignancies. Haematologica 2022;107(6):1243–1263. doi: 10.3324/haematol.2021.279512
    • 40. National Institutes of Health National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. Washington (DC): United States Department of Health and Human Services. [updated 2017 Nov 27; cited 2024 Feb 16]. Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5x11.pdf
    • 41. Crawford J, Moore DC, Morrison VA et al. Use of prophylactic pegfilgrastim for chemotherapy-induced neutropenia in the US: a review of adherence to present guidelines for usage. Cancer Treat Res. Commun. 2021;29:100466. doi: 10.1016/j.ctarc.2021.100466
    • 42. Stephens JM, Li X, Reiner M et al. Annual patient and caregiver burden of oncology clinic visits for granulocyte-colony stimulating factor therapy in the US. J. Med. Econ. 2016;19(5):537–547. doi: 10.3111/13696998.2016.1140052
    • 43. Fortner BV, Tauer KW, Okon T et al. Experiencing neutropenia: quality of life interviews with adult cancer patients. BMC Nurs. 2005;4:4. doi: 10.1186/1472-6955-4-4
    • 44. Bryer E, Henry D. Chemotherapy-induced anemia: etiology, pathophysiology, and implications for contemporary practice. Int. J. Clin. Transfus. 2018;6:21–31. doi: 10.2147/IJCTM.S187569
    • 45. Butt Z, Rosenbloom SK, Abernethy AP et al. Fatigue is the most important symptom for advanced cancer patients who have had chemotherapy. J. Natl Compr. Canc. Netw. 2008;6(5):448–455. doi: 10.6004/jnccn.2008.0036
    • 46. Corey-Lisle PK, Desrosiers MP, Collins H et al. Transfusions and patient burden in chemotherapy-induced anaemia in France. Ther. Adv. Med. Oncol. 2014;6(4):146–153. doi: 10.1177/1758834014534515 • A retrospective chart review of patients with cancer receiving an outpatient red blood cell (RBC) transfusion in France, which demonstrated that RBC transfusion is a burden for patients in terms of time spent in the clinic and the associated travel. The article also references other work showing that transportation costs, lost wages and other factors may also contribute to the patient burden.
    • 47. Cancer.Net. Low platelet count or thrombocytopenia. Alexandria (VI): American Society of Clinical Oncology (ASCO); c2005-2024. [updated 2020 Aug; cited 2024 Feb 16]. Available from: www.cancer.net/coping-with-cancer/physical-emotional-and-social-effects-cancer/managing-physical-side-effects/low-platelet-count-or-thrombocytopenia
    • 48. FACIT.org. FACIT Measures & Languages. Ponte Vedra Beach (FL): FACIT.org; c2001. [updated 2021; cited 2024 Feb 16]. Available from: www.facit.org/measures-language-availability
    • 49. Vadhan-Raj S. Management of chemotherapy-induced thrombocytopenia: current status of thrombopoietic agents. Semin. Hematol. 2009;46(2 Suppl. 1):S26–S32. doi: 10.1053/j.seminhematol.2008.12.007
    • 50. Lyman GH, Abella E, Pettengell R. Risk factors for febrile neutropenia among patients with cancer receiving chemotherapy: a systematic review. Crit. Rev. Oncol. Hematol. 2014;90(3):190–199. doi: 10.1016/j.critrevonc.2013.12.006
    • 51. Cupp J, Culakova E, Poniewierski MS et al. Analysis of factors associated with in-hospital mortality in lung cancer chemotherapy patients with neutropenia. Clin. Lung Cancer 2018;19(2):e163–e169. doi: 10.1016/j.cllc.2017.10.013
    • 52. Okon T, Fortner B, Schwartzberg L et al. Quality of life (QOL) in patients with grade IV chemotherapy-induced neutropenia (CIN) [abstract 2920]. Proc. Am. Soc. Clin. Oncol. 2002;21:275b.
    • 53. Epstein RS, Basu Roy UK, Aapro M et al. Cancer patients' perspectives and experiences of chemotherapy-induced myelosuppression and its impact on daily life. Patient Prefer. Adher. 2021;15:453–465. doi: 10.2147/PPA.S292462
    • 54. Kristensen A, Solheim TS, Fløtten Ø et al. Associations between hematologic toxicity and health-related quality of life during first-line chemotherapy in advanced non-small-cell lung cancer: a pooled analysis of two randomized trials. Acta Oncol. 2018;57(11):1574–1579. doi: 10.1080/0284186X.2018.1492151
    • 55. Wagner LI, Beaumont JL, Ding B et al. Measuring health-related quality of life and neutropenia-specific concerns among older adults undergoing chemotherapy: validation of the Functional Assessment of Cancer Therapy-Neutropenia (FACT-N). Support. Care Cancer 2008;16(1):47–56. doi: 10.1007/s00520-007-0270-7
    • 56. Wang XJ, Wong CM, Chan A. Psychometric properties of the functional assessment of cancer therapy-neutropenia in asian cancer patients with chemotherapy-induced neutropenia. J. Pain Symptom Manage. 2016;52(3):428–436. doi: 10.1016/j.jpainsymman.2016.03.016
    • 57. Bendall LJ, Bradstock KF. G-CSF: from granulopoietic stimulant to bone marrow stem cell mobilizing agent. Cytokine Growth Factor Rev. 2014;25(4):355–367. doi: 10.1016/j.cytogfr.2014.07.011
    • 58. Link H. Current state and future opportunities in granulocyte colony-stimulating factor (G-CSF). Support. Care Cancer 2022;30(9):7067–7077. doi: 10.1007/s00520-022-07103-5
    • 59. 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. 2013;24(10):2475–2484. doi: 10.1093/annonc/mdt226
    • 60. Lambertini M, Del Mastro L, Bellodi A et al. The five “Ws” for bone pain due to the administration of granulocyte-colony stimulating factors (G-CSFs). Crit. Rev. Oncol. Hematol. 2014;89(1):112–128. doi: 10.1016/j.critrevonc.2013.08.006 • This review article discusses the incidence, causes, consequences and management of bone pain in cancer patients receiving granulocyte colony-stimulating factors.
    • 61. Macmillan Cancer Support. G-CSF (granulocyte-colony stimulating factor). London, United Kingdom: Macmillan Cancer Support; c2024. [updated 2024; cited 2024 Feb 16]. Available from: www.macmillan.org.uk/cancer-information-and-support/treatments-and-drugs/g-csf
    • 62. United States Food and Drug Administration. NEULASTA® (pegfilgrastim) injection [patient information]. Thousand Oaks (CA): Amgen, Inc.; January 2021 [cited 2024 Feb 16]. Available from: www.pi.amgen.com/-/media/Project/Amgen/Repository/pi-amgen-com/Neulasta/neulasta_ppi_pt_english.pdf
    • 63. United States Food and Drug Administration. ROLVEDON™ (eflapegrastim-xnst) injection, for subcutaneous use [prescribing information]. Irvine (CA): Spectrum Pharmaceuticals, Inc.; June 2023 [cited 2024 Feb 16]. Available from: www.rolvedon.com/pdf/rolvedon-prescribing-information.pdf
    • 64. Crawford J, Oswalt C. The impact of new and emerging agents on outcomes for febrile neutropenia: addressing clinical gaps. Curr. Opin. Oncol. 2023;35(4):241–247. doi: 10.1097/CCO.0000000000000952
    • 65. Saleh MN, Patel MR, Bauer TM et al. Phase I trial of ALRN-6924, a dual inhibitor of MDMX and MDM2, in patients with solid tumors and lymphomas bearing wild-type TP53. Clin. Cancer Res. 2021;27(19):5236–5247. doi: 10.1158/1078-0432.CCR-21-0715
    • 66. Guerlavais V, Sawyer TK, Carvajal L et al. Discovery of sulanemadlin (ALRN-6924), the first cell-permeating, stabilized α-helical peptide in clinical development. J. Med. Chem. 2023;66(14):9401–9417. doi: 10.1021/acs.jmedchem.3c00623
    • 67. OncLive®. Aileron terminates Phase Ib trial evaluating ALRN-6924 in P53-mutant breast cancer. Cranbury (NJ): MJH Life Sciences. [updated 2023 Feb 22; cited 2024 Feb 16]. Available from: www.onclive.com/view/aileron-terminates-phase-1b-trial-evaluating-alrn-6924-in-p53-mutant-breast-cancer
    • 68. Bekes I, Eichler M, Singer S et al. Impact of granulocyte colony-stimulating factor (G-CSF) and epoetin (EPO) on hematologic toxicities and quality of life in patients during adjuvant chemotherapy in early breast cancer: results from the multi-center randomized ADEBAR trial. Clin. Breast Cancer 2020;20(6):439–447. doi: 10.1016/j.clbc.2020.03.008
    • 69. Kenar G, Köksoy EB, Ürün Y et al. Prevalence, etiology and risk factors of anemia in patients with newly diagnosed cancer. Support. Care Cancer 2020;28(11):5235–5242. doi: 10.1007/s00520-020-05336-w
    • 70. Crawford J, Kosmidis PA, Hirsch FR et al. Targeting anemia in patients with lung cancer. J. Thorac. Oncol. 2006;1(7):716–725. doi: 10.1016/S1556-0864(15)30388-9
    • 71. Lloyd A, van Hanswijck de Jonge P, Doyle S et al. Health state utility scores for cancer-related anemia through societal and patient valuations. Value Health 2008;11(7):1178–1185. doi: 10.1111/j.1524-4733.2008.00394.x
    • 72. LIVESTRONG “I learned to live with it” is not good enough: Challenges reported by post-treatment cancer survivors in the LIVESTRONG surveys. A LIVESTRONG report, 2010 Austin (TX): The Livestrong Foundation. [updated 2011; cited 2024 Feb 16]. Available from: https://www.livestrong.org/sites/default/files/what-we-do/reports/lssurvivorsurveyreport_final_0.pdf
    • 73. Kink P, Egger EM, Lanser L et al. Immune activation and anemia are associated with decreased quality of life in patients with solid tumors. J. Clin. Med. 2020;9(10):3248. doi: 10.3390/jcm9103248
    • 74. Cella D, Lai JS, Stone A. Self-reported fatigue: one dimension or more? Lessons from the Functional Assessment of Chronic Illness Therapy–Fatigue (FACIT-F) questionnaire. Support. Care Cancer 2011;19(9):1441–1450. doi: 10.1007/s00520-010-0971-1
    • 75. Crawford J, Cella D, Cleeland CS et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002;95(4):888–895. doi: 10.1002/cncr.10763
    • 76. Cella D, Kallich J, McDermott A et al. The longitudinal relationship of hemoglobin, fatigue and quality of life in anemic cancer patients: results from five randomized clinical trials. Ann. Oncol. 2004;15(6):979–986. doi: 10.1093/annonc/mdh235
    • 77. Littlewood TJ, Bajetta E, Nortier JW et al. Effects of epoetin alfa on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: results of a randomized, double-blind, placebo-controlled trial. J. Clin. Oncol. 2001;19(11):2865–2874. doi: 10.1200/JCO.2001.19.11.2865
    • 78. Bokemeyer C, Aapro MS, Courdi A et al. EORTC guidelines for the use of erythropoietic proteins in anaemic patients with cancer: 2006 update. Eur. J. Cancer 2007;43(2):258–270. doi: 10.1016/j.ejca.2006.10.014
    • 79. Mäenpää J, Puistola U, Riska H et al. Impact of epoetin-beta on anemia and health-related quality of life in cancer patients: a prospective observational study using the generic 15D instrument. Anticancer Res. 2014;34(5):2325–2329.
    • 80. Pronzato P, Cortesi E, van der Rijt CC et al. Epoetin alfa improves anemia and anemia-related, patient-reported outcomes in patients with breast cancer receiving myelotoxic chemotherapy: results of a European, multicenter, randomized, controlled trial. Oncologist 2010;15(9):935–943. doi: 10.1634/theoncologist.2009-0279
    • 81. Straus DJ, Testa MA, Sarokhan BJ et al. Quality-of-life and health benefits of early treatment of mild anemia: a randomized trial of epoetin alfa in patients receiving chemotherapy for hematologic malignancies. Cancer 2006;107(8):1909–1917. doi: 10.1002/cncr.22221
    • 82. Gabrilove JL, Perez EA, Tomita DK et al. Assessing symptom burden using the M. D. Anderson symptom inventory in patients with chemotherapy-induced anemia: results of a multicenter, open-label study (SURPASS) of patients treated with darbepoetin-alpha at a dose of 200 microg every 2 weeks. Cancer 2007;110(7):1629–1640. doi: 10.1002/cncr.22943
    • 83. Vega A, Zhang R, Wong HL et al. Trends in erythropoiesis-stimulating agent use and blood transfusions for chemotherapy-induced anemia throughout FDA's risk evaluation and mitigation strategy lifecycle. Pharmacoepidemiol. Drug Saf. 2021;30(5):626–635. doi: 10.1002/pds.5202
    • 84. Schoen MW, Hoque S, Witherspoon BJ et al. End of an era for erythropoiesis-stimulating agents in oncology. Int. J. Cancer 2020;146(10):2829–2835. doi: 10.1002/ijc.32917
    • 85. Sarpatwari A, He M, Tessema FA et al. Changes in erythropoiesis stimulating agent use under a Risk Evaluation and Mitigation Strategy (REMS) program. Drug Saf. 2021;44(3):327–335. doi: 10.1007/s40264-020-01017-z
    • 86. Weycker D, Hatfield M, Grossman A et al. Risk and consequences of chemotherapy-induced thrombocytopenia in US clinical practice. BMC Cancer 2019;19(1):151. doi: 10.1186/s12885-019-5354-5
    • 87. Kuter DJ. Managing thrombocytopenia associated with cancer chemotherapy. Oncology (Williston Park) 2015;29(4):282–294.
    • 88. Cella D, Beaumont JL, Webster KA et al. Measuring the concerns of cancer patients with low platelet counts: The Functional Assessment of Cancer Therapy–thrombocytopenia (FACT-Th) questionnaire. Support. Care Cancer 2006;14(12):1220–1231. doi: 10.1007/s00520-006-0102-1
    • 89. Kaufman RM, Djulbegovic B, Gernsheimer T et al. Platelet transfusion: a clinical practice guideline from the AABB. Ann. Intern. Med. 2015;162(3):205–213. doi:10.7326/M14-1589
    • 90. Kiefel V. Reactions induced by platelet transfusions. Transfus Med. Hemother. 2008;35(5):354–358. doi: 10.1159/000151350
    • 91. George JN, Mathias SD, Go RS et al. Improved quality of life for romiplostim-treated patients with chronic immune thrombocytopenic purpura: results from two randomized, placebo-controlled trials. Br. J. Haematol. 2009;144(3):409–415. doi: 10.1111/j.1365-2141.2008.07464.x