This site is intended for healthcare professionals
Doctor holding patient
Supportive care in oncology Learning Zone

Supportive care options for cancer

Read time: 80 mins
Last updated:19th May 2024
Published:25th Aug 2022

Managing side effects associated with chemotherapy and radiation therapy

Chemotherapy drugs are used extensively in clinic as one of the main cancer treatments and can be administered alone or as adjuvant therapy. Chemotherapy interferes with and inhibits DNA, RNA and protein synthesis, often in the S phase of the cell cycle, leading to cell death. Radiation therapy is commonly used in conjunction with chemotherapy and has a similar mode of action, causing damage to DNA strands and resulting in cell death1.

Although chemotherapy and radiation therapy have improved the survival of people with cancer, they are notorious for causing severe side effects that ultimately limit the amount that can be tolerated and, thus, treatment efficacy1

Drugs commonly used for chemotherapy include1:

  • Platinum-based agents (cisplatin, oxaliplatin, carboplatin)
  • Taxanes (paclitaxel, docetaxel)
  • Fluoropyrimidines (5-fluorouracil, capecitabine)
  • Vinca alkaloids (vinblastine, vinorelbine, vincristine, vindesine)  

Guideline recommendations for managing the side effects of chemotherapy and radiation therapy

Fatigue

View transcript

Management of cancer-related fatigue includes an initial screening for contributing factors such as sleep disturbance and anaemia2. Management interventions include exercise (a mixture of aerobic, strength and flexibility exercises), cognitive behavioural therapy, and pharmacological management with psychostimulants such as methylphenidate and dexamphetamine2.

Thrombosis

Dr Lyman discusses the challenges of treating thrombosis in people with cancer. View transcript

Venous thromboembolism (VTE) is a major complication of cancer. Anticoagulants are used for VTE prevention, and treatment options include3-8:

  • Unfractionated heparin (UFH)
  • Low-molecular-weight heparins (LMWHs)
  • Fondaparinux (a synthetic indirect inhibitor of activated factor Xa)
  • Vitamin K antagonists (VKAs)
  • Direct oral anticoagulants (DOACS)

Choice of anticoagulant is determined by considerations such as bleeding risk, patient preference and potential drug interactions4-8. LMWH is the preferred treatment choice in those at high risk of bleeding, while for patients at low risk of bleeding, DOACs (apixaban or rivaroxaban) are preferred4-8. Caution should be exercised when prescribing DOACS to people with gastrointestinal or genitourinary tract cancers because of an increased risk of bleeding4,5,7,8. If apixaban, rivaroxaban or LMWHs are contraindicated or unsuitable, alternatives such as dabigatran, UFH, fondaparinux or a VKA can be considered5.

For symptomatic or incidental VTE, treatment is recommended for at least 3–6 months, or for as long as the cancer is active, the patient remains on cancer therapy or remains at high risk of recurrent VTE5,6.

The American Society of Hematology (ASH) 2021 guidelines suggest indefinite anticoagulant treatment for the long-term prevention of VTE in patients with cancer may be considered if the benefits outweigh the risks5. Similarly, the European Society for Medical Oncology (ESMO) 2023 guidelines and American Society of Clinical Oncology (ASCO) 2023 guidelines suggest extending anticoagulant treatment beyond 6 months in patients in whom the risk of recurrence outweighs the risks of bleeding complications, and that the risk–benefit profile should be regularly reassessed6,9.

More on the management of cancer-associated thrombosis

Chemotherapy-induced thrombocytopenia

The National Comprehensive Cancer Network (NCCN) 2024 guidelines recommend considering platelet transfusion, chemotherapy dose reduction or regimen change, or a thrombopoietin receptor agonist (TPO-RA) within a clinical trial as treatment10. The International Society on Thrombosis and Haemostasis (ISTH) similarly recommend platelet transfusion for severe chemotherapy-induced thrombocytopenia (CIT) or a TPO-RA as part of a clinical trial for CIT in people with solid tumours, with the goal of TPO-RA use to achieve an adequate platelet count and avoid reductions in chemotherapy dose intensity in future cycles11.

Platelet transfusion remains the cornerstone of managing CIT; however, potential problems including allogeneic immunity, infectious pathogen transfer and transfusion reactions need to be considered11,12. A synthesis of the available data on management practices is as follows10,11,13,14:

  • Identify and treat any other underlying cause of thrombocytopenia (stop antibiotics, treat infections, and control coagulopathy)
  • Reduce chemotherapy dose, frequency or alter the chemotherapy regimen
  • Platelet transfusion support can be used if maintenance of chemotherapy dose intensity is vital for response or survival, and is indicated if the patient is bleeding or to prevent major bleeding if platelet counts are less than 10 × 109/L (<20 × 109/L if febrile)
  • Recombinant interleukin-11 (oprelvekin) is the only approved treatment for CIT; however, its use is limited by adverse effects
  • TPO-RAs increase platelet production but are not approved for CIT, and their use should ideally be as part of a clinical trial; if TPO-RAs are considered, romiplostim is the preferred agent
  • Antifibrinolytic agent tranexamic acid is not recommended for the prevention of haemorrhage in CIT

Febrile and non-febrile neutropenia

Dr Lyman discusses guideline-recommended management of febrile and non-febrile neutropenia following cancer treatment. View transcript

Most patients undergoing chemotherapy and radiation therapy will develop neutropenia, which is defined by the absolute neutrophil count (ANC) and categorised as moderate (ANC <1,000 cells/µL), severe (ANC <500 cells/µL) or profound (ANC <100 cells/µL)15.

Patients with neutropenia are highly susceptible to infection and require careful monitoring for fever, chills and sweats15

The most dangerous complication of neutropenia is febrile neutropenia (FN), which is defined by ESMO and NCCN guidelines as a one-time oral temperature of >38.3°C or a sustained temperature of >38°C (100.4°F) for ≥1 hour in a patient who has an ANC of <500 cells/μL10,16. ASCO guidelines define FN as a one-time oral temperature of >38.3°C or a sustained temperature of >38°C (100.4°F) for ≥1 hour in a patient who has an ANC of <1,000 cells/μL15,17.

FN can be effectively prevented by administering granulocyte colony-stimulating factors (G-CSFs); meta-analyses indicate that primary prophylaxis with G-CSF (filgrastim) reduces the risk of FN by at least 50%18,19. However, G-CSF prophylaxis is recommended only for patients who are to receive a chemotherapy regimen with a high FN risk (≥20%) or who have an overall FN risk ≥20%10,16,20. Overall FN risk is estimated for patients who receive a chemotherapy regimen with an intermediate FN risk (10–20%). For these patients, special attention is given to patient-related risk factors, such as age >65 years and the presence of serious comorbidities. G-CSF prophylaxis is not indicated if the planned chemotherapy regimen has a low FN risk (<10%) or if overall FN risk is <20%.10,16,20.

Age ≥65 years, an Eastern Cooperative Oncology Group Score ≥2 and advanced stage (≥2) are risk factors for FN, and the level of risk is also influenced by the type of cancer, cytotoxic regimen, dose intensity, comorbidities and previous episodes of FN10,16,17,20.

Initial management should include taking a detailed medical history, including past infections with antibiotic-resistant organisms. A full-body examination looking for infection entry points should be performed, as well as routine investigations, including16,21:

  • Urgent blood testing to assess bone marrow, renal and liver function
  • Coagulation screen
  • C-reactive protein
  • Blood cultures, including cultures from indwelling intravenous catheters
  • Urinalysis and culture
  • Sputum microscopy and culture
  • Stool microscopy and culture
  • Skin lesion assessment (aspirate/biopsy/swab)
  • Chest radiograph

The majority of FN cases can be resolved with prompt empirical therapy with tailored antibacterial therapy if the type of infection is known16. In patients at high risk of FN, treatment with intravenous broad-spectrum antibiotics is recommended16,21. Oral antibacterial therapy can be started for patients at low risk of FN who16,21:

  • Are haemodynamically stable
  • Do not have acute leukaemia or evidence of organ failure
  • Do not have pneumonia, an indwelling venous catheter or severe soft tissue infection

ESMO guidelines recommend using the Multinational Association of Supportive Care in Cancer (MASCC) FN risk calculator to pre-emptively manage this condition16. A MASCC score of ≤20 indicates high risk, while a score of ≥21 indicates low risk22. ESMO guidelines state that patients identified as high risk by the MASCC criteria should be admitted to hospital and receive inpatient antibiotic therapy, whereas those at low risk may be treated with outpatient parenteral regimens16.

However, ASCO and the Infectious Diseases Society of America (IDSA) guidelines recommend using clinical judgement criteria in addition to tools such as the MASCC criteria, Talcott’s rules, or Clinical Index of Stable Febrile Neutropenia (CISNE) to decide if patients with FN require inpatient or outpatient care (Figure 1)15.

Figure 1. Algorithm for the management of febrile neutropenia in patients treated for malignancy15,16. CISNE, Clinical Index of Stable Febrile Neutropenia; MASCC, Multinational Association of Supportive Care in Cancer.

Clinical judgement criteria that can that indicate the need for inpatient care may include accelerated hypertension, severe thrombocytopenia, inability to swallow oral medications, impaired hepatic function and an altered mental status15. Talcott’s rules classify patients into groups 1, 2, 3 or 4. Patients in groups 1–3 are considered to be at high risk and include those who were hospitalised at the time of fever onset; outpatients with an acute comorbidity requiring hospitalisation; and outpatients without comorbidities but with uncontrolled cancer. Talcott’s group 4 comprises outpatients without comorbidities and with controlled cancer, who are considered to be at low risk15.

In a pooled analysis, the MASCC score and Talcott’s rules were found to misclassify some patients as being at low risk, and serious complications developed in ≤11% of patients classified as being at low risk by the MASCC score and in 7% of patients in Talcott’s group 415. Hence, ASCO/IDSA guidelines also recommended use of the CISNE tool, which has demonstrated better performance characteristics than the MASCC criteria and Talcott’s rules, and can improve classification of patients with solid tumours. The CISNE tool considers six variables integrated into a score that deems a score of 0 as low risk, a score of 1–2 points as intermediate risk, and a score of ≥3 as high risk. Patients with a CISNE score of ≥3 are candidates for inpatient management15.

The use of antimicrobials to prevent FN is discouraged by ESMO, but is recommended by ASCO/IDSA for patients at high risk of infection, such as patients expected to have profound, protracted neutropenia (<100 neutrophils/µL  for >7 days)16,17.

Monotherapy and combination therapy have equivalent efficacy; however, patients at high risk of FN may benefit more from a β-lactam antibiotic in combination with an aminoglycoside15,16. Clinical assessment and follow-up may be required every 2–4 hours in severe cases, while daily assessment of fever trends, bone marrow and renal function is recommended until the patient is afebrile for at least 24 hours16.

Nausea and vomiting

Dr Lyman highlights guideline-recommended management strategies for nausea and vomiting caused by cancer treatments. View transcript.

The risk of chemotherapy-induced nausea and vomiting (CINV) for each patient should be ascertained on the basis of emetogenic potential of the treatment (Table 1 and Table 2) and predisposing factors, such as gender, type of cancer and age. A useful tool recommended by the MASCC and ESMO for predicting CINV risk can be found at www.riskcinv.com.

Table 1. Emetogenic risk of intravenous anticancer treatments23,24.

Emetic risk Definition of risk Intravenous agents
High Risk in nearly all patients (>90%) • Anthracycline/cyclophosphamide combination 
• Carmustine 
• Chlormethine (mechlorethamine) 
• Cisplatin
• Cyclophosphamide ≥1500 mg/m2 
• Dacarbazine
• Streptozocin
Moderate Risk in 30% to 90% of patients • Alemtuzumab
• Arsenic trioxide
• Azacitidine
• Bendamustine
• Busulfan
• Carboplatin
• Clofarabine
• Cyclophosphamide <1500 mg/m2
• Cytarabine >1000 mg/m2
• Cytarabine/daunorubicin liposomal
• Daunorubicin
• Dinutuximab beta
• Doxorubicin
• Epirubicin Idarubicin
• Ifosfamide
• Irinotecan
• Irinotecan peg-liposomal
• Lurbinectedin
• Naxitamab
• Oxaliplatin
• Romidepsin
• Sacituzumab-govitecan
• Temozolomide
• Thiotepa
• Trabectedin
• Trastuzumab-deruxtecan
Low Risk in 10% to 30% of patients  • Aflibercept
• Amivantamab
• Axicabtagene-ciloleucel
• Belinostat
• Blinatumomab
• Bortezomib
• Brentuximab-vedotin
• Cabazitaxel
• Carfilzomib
• Catumaxomab
• Cetuximab
• Copanlisib
• Cytarabine ≤1000 mg/m2
• Decitabine
• Docetaxel
• Doxorubicin peg-liposomal
• Elotuzumab
• Enfortumab-vedotin
• Eribulin
• Etoposide
• 5-Fluorouracil
• Gemcitabine
• Gemtuzumab-ozogamicin
• Inotuzumab-ozogamicin
• Isatuximab
• Ixabepilone
• Loncastuximab-tesirine
• Margetuximab
• Melphalan-flufenamide
• Methotrexate
• Mirvetuximab-soravtansine
• Mitomycin
• Mitoxantrone
• Moxetumomab-pasudotox
• Necitumumab
• Nelarabine
• Paclitaxel
• Paclitaxel nab-albumin
• Panitumumab
• Pemetrexed
• Pertuzumab
• Tafasitamab
• Tagraxofusp
• Teclistamab
• Temsirolimus
• Tisagenlecleucel
• Tisotumab-vedotin
• Topotecan
• Trastuzumab-emtansine
• Vinflunine
Minimal Fewer than 10% at risk  • Asparaginase
• Atezolizumab
• Avelumab
• Belantamab-mafodotin
• Bevacizumab
• Bleomycin
• Cemiplimab
• Cladribine (2-chlorodeoxyadenosine)
• Daratumumab
• Dostarlimab
• Durvalumab
• Emapalumab
• Fludarabine
• Ipilimumab
• Mosunetuzumab
• Nivolumab
• Obinutuzumab
• Ofatumumab
• Pembrolizumab
• Pixantrone
• Polatuzumab-vedotin
• Pralatrexate
• Ramucirumab
• Rituximab
• Trastuzumab
• Tremelimumab
• Vinblastine
• Vincristine
• Vinorelbine

Table 2. Emetogenic risk of oral anticancer treatments23.

Emetic risk Definition of risk Oral agents
High/moderate Risk in more than 30% of patients • Abemaciclib
• Adagrasib
• Avapritinib
• Bosutinib
• Cabozantinib
• Ceritinib
• Crizotinib
• Cyclophosphamide
• Enasidenib
• Fedratinib
• Hexamethylmelamine (altretamine)
• Imatinib
• Lenvatinib
• Lomustine
• Midostaurin
• Mobocertinib
• Niraparib
• Olaparib
• Procarbazine
• Ribociclib
• Rucaparib
• Selinexor
• Temozolomide
• Vinorelbine
Low/minimal Risk in fewer than 30% of patients • Acalabrutinib
• Afatinib
• Alectinib
• Alpelisib
• Apalutamide
• Asciminib
• Axitinib
• Bexarotene
• Brigatinib
• Capecitabine
• Capmatinib
• Chlorambucil
• Cobimetinib
• Dabrafenib
• Dacomitinib
• Darolutamide
• Dasatinib
• Duvelisib
• Encorafenib
• Entrectinib
• Erdafitinib
• Erlotinib
• Estramustine
• Etoposide
• Everolimus
• Fludarabine
• Futibatinib
• Gefitinib
• Gilteritinib
• Glasdegib
• Hydroxyurea
• Ibrutinib
• Idelalisib
• Infigratinib
• Ivosidenib
• Ixazomib
• Lapatinib
• Larotrectinib
• Lenalidomide
• Lorlatinib
• Melphalan (L-Phenylalanine mustard)
• Methotrexate
• Neratinib
• Nilotinib
• Nintedanib
• Olutasidenib
• Osimertinib
• Palbociclib
• Panobinostat
• Pazopanib
• Pemigatinib
• Pexidartinib
• Pomalidomide
• Ponatinib
• Pralsetinib
• Regorafenib
• Relugolix
• Ripretinib
• Ruxolitinib
• Selpercatinib
• Sonidegib
• Sorafenib
• Sotorasib
• Sunitinib
• Talazoparib
• Tazemetostat
• Tegafur/uracil
• Tepotinib
• Thalidomide
• Tioguanin (6-thioguanine)
• Tivozanib
• Topotecan
• Trametinib
• Trifluridine/tipiracil
• Tucatinib
• Umbralisib
• Vandetanib
• Vemurafenib
• Venetoclax
• Vismodegib
• Vorinostat
• Zanubrutinib

Management of CINV depends on the emetogenic potential of the anticancer therapy that a patient is taking (high risk: emesis documented in >90% of patients; moderate risk: emesis documented in 30–90% of patients; low risk: emesis documented in 10–30% of patients; minimal risk: emesis documented in <10% of patients)23-25.

High emetic risk

European and US guidelines recommend a four-drug combination of a neurokinin-1 (NK1) receptor antagonist, a 5-hydroxytryptamine 3 (5-HT3) receptor antagonist (ondansetron, granisetron), dexamethasone and olanzapine on day 1 of treatment to prevent acute nausea and vomiting. Dexamethasone and olanzapine should be continued on days 2–4 to prevent delayed nausea and vomiting in patients on anthracycline/cyclophosphamide (AC) chemotherapy24,25. For patients on non-AC chemotherapy, olanzapine should be continued on days 2–4 to prevent delayed nausea and vomiting24,25.

For high-emetic-risk total body radiation therapy, prophylaxis with a two-drug combination of a 5-HT3 receptor antagonist and dexamethasone is recommended24,25

Moderate emetic risk

Adults treated with carboplatin-based moderate-emetic-risk treatments should be offered a three-drug regimen of a 5-HT3 receptor antagonist, dexamethasone and an NK1 receptor antagonist before treatment to prevent acute nausea and vomiting24,25. For oxaliplatin-based or other non-carboplatin-based moderate-emetic-risk treatments, patients should be offered a two-drug combination of a 5-HT3 receptor antagonist (preferably palonosetron) and dexamethasone before treatment. An NK1 receptor antagonist may be added to the regimen for women ≤50 years of age receiving oxaliplatin-based chemotherapy24,25. ASCO further states that patients treated with cyclophosphamide, doxorubicin, oxaliplatin and other moderate-emetic-risk drugs known to cause delayed CINV may be offered dexamethasone on days 2–325.

Patients receiving upper abdomen/craniospinal moderate-emetic-risk radiation therapy should be offered prophylaxis with a 5-HT3 receptor antagonist with optional dexamethasone24,25.

Low emetic risk

European and US guidelines recommend that patients treated with low-emetic-risk agents may be offered a single antiemetic agent, such as a 5-HT3 receptor antagonist or dexamethasone, before treatment for the prevention of acute nausea and vomiting24,25. European guidelines also suggest a dopamine receptor antagonist, such as metoclopramide, as an option24. Patients should not be routinely offered an antiemetic agent prior to receiving minimal-risk emetic treatments24,25. Patients should also not be routinely offered an antiemetic for the prevention of delayed nausea and vomiting following low or minimal-emetic-risk treatments24.

Patients undergoing low-emetic-risk radiation therapy to the brain can be offered rescue dexamethasone therapy24,25. Patients who are treated with low-emetic-risk radiation therapy to the head and neck, thorax or pelvis, or minimal-risk radiation therapy to the extremities or breast can be offered rescue therapy with either dexamethasone, a 5-HT3 receptor antagonist, or a dopamine-receptor antagonist24,25.

Mucositis

Oral and gastrointestinal (GI) mucositis caused by high-dose chemotherapy and/or radiation continues to be an important clinical problem. The ESMO 2015 guidelines recommend the following for the prevention and treatment of oral mucositis26:

  • 30 minutes of oral cryotherapy to prevent oral mucositis in patients receiving bolus 5-fluorouracil chemotherapy
  • Recombinant human keratinocyte growth factor-1 (KGF-1/palifermin) to prevent oral mucositis in patients receiving high-dose chemotherapy and total body irradiation followed by haematopoietic stem cell transplantation
  • Low-level laser therapy to prevent oral mucositis in patients receiving haematopoietic stem cell transplantation conditioned with high-dose chemotherapy, with or without total body irradiation
  • Patient-controlled analgesia with morphine to treat pain due to oral mucositis
  • Benzydamine mouthwash to prevent oral mucositis in patients with head and neck cancer receiving moderate-dose radiation therapy (up to 50 Gy) without concomitant chemotherapy

The MASCC and International Society of Oral Oncology (ISOO) support these recommendations and additionally recommend low-level laser therapy for the prevention of oral mucositis in adults receiving radiation therapy to the head and neck, with or without chemotherapy27. For patients undergoing haematopoietic stem cell transplantation, oral cryotherapy is recommended when conditioning includes high-dose melphalan, and KGF-1 is recommended when conditioning includes high-dose chemotherapy and total body irradiation27.

The ESMO 2015 guideline recommendations for the prevention and treatment of GI mucositis include26:

  • The use of intravenous amifostine in patients receiving radiation therapy to prevent radiation proctitis
  • Subcutaneous octreotide twice daily to treat diarrhoea secondary to mucositis induced by standard- or high-dose chemotherapy if loperamide is ineffective  

Diarrhoea

Diarrhoea is a common side effect of various antineoplastic treatments and can be classified as uncomplicated or complicated28.

To ascertain the severity of diarrhoea, it should first be graded using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE)29

Uncomplicated diarrhoea

Grade 1 or 2 diarrhoea with no other complications may be classified as ‘uncomplicated’ and managed conservatively with oral hydration and loperamide (4 mg to start and then 2 mg every 4 hours or after each loose stool)28. Skin barrier creams should be used to prevent skin irritation caused by increased faecal contact28.

Complicated diarrhoea

Patients with complicated diarrhoea, which includes mild-to-moderate diarrhoea accompanied by moderate-to-severe cramping, nausea and vomiting, cognitive decline, fever, sepsis, neutropenia, bleeding or dehydration, require hospitalisation for closer monitoring and evaluation28.

Complicated diarrhoea is managed with intravenous rehydration fluids, antibiotics and subcutaneous octreotide at a starting dose of 100–150 µg three times a day. If the patient is severely dehydrated, octreotide can be administered intravenously at a dose of 25–50 µg/h. The dose can be rapidly escalated up to 500 µg subcutaneously until the diarrhoea is controlled. Additional evaluations to be performed include a complete blood count, electrolyte profile and a stool workup investigating the presence of blood, Clostridium difficile, Salmonella, Escherichia coli, Campylobacter and infectious colitis28.

Constipation

Best practice for management of constipation is a balance between prevention, self-care, and oral and rectal laxative therapy. Self-care strategies include increasing fluids, dietary fibre intake and mobility30.

Other classes of laxatives, including bulk-forming laxatives, detergent/stool softener and liquid paraffin, are generally not recommended over osmotic and stimulant laxatives, especially in cases of advanced cancer30.

Suppositories and enemas are the recommended first-line therapy where there is faecal impaction and/or where oral laxatives have failed to relieve constipation30.

Acute radiation dermatitis

A standardised approach to management of acute radiation dermatitis (ARD) has been limited by the lack of high-quality evidence. To address this gap, in January 2023, MASCC conducted a systematic review of current evidence on interventions for the prevention and treatment of ARD31. This review included 235 studies, of which 149 were randomised controlled clinical trials; however, definitive recommendations could not be made on the basis of published evidence as there was considerable variability and insufficient high-quality evidence. A four-round Delphi consensus process was therefore undertaken to generate recommendations based on the opinions of 42 international ARD experts. Interventions were recommended if at least 75% consensus was reached32.

The Delphi consensus process recommended six interventions for the prevention of ARD, including photobiomodulation therapy and silicone film (in people with breast cancer), polyurethane film, mometasone furoate, betamethasone and olive oil. For the management of ARD, foam dressings were recommended (Table 3)32.

The study concluded that most interventions could not be recommended because there was insufficient evidence, conflicting evidence, or lack of consensus to support use32. This suggests the need for further research on the prevention and management of ARD. However, clinicians can consider implementing the recommended interventions in their practice until additional evidence becomes available.

Table 3. Delphi consensus recommendations for interventions to prevent or manage acute radiation dermatitis32. Note, recommended interventions are those with a consensus of ≥75%. Interventions with consensus of <75% could not be recommended.

Intervention Consensus to recommend
Prevention
Laser therapy
• Photobiomodulation or low-level laser therapy (breast cancer)
Barrier films and dressings
• Silicone-based polyurethane (breast cancer)
• Polyurethane film
Topical corticosteroids
• Mometasone
• Betamethasone
Natural and miscellaneous agents
• Olive oil
• Nigella sativa extract
Topical non-steroidal agents
• Aqueous cream
• Hyaluronic acid
• Heparinoid
Antibiotics
• Sulfadiazine silver
Growth factors and other oral agents
• Epidermal growth factors
Antiperspirant or deodorant
• Non-aluminium or non-metallic


79%

76%
94%

94%
97%

79%
45%

52%
39%
36%

73%

6%

21%
Management
Barrier films and dressings
• Foam dressing
• Silicone-based polyurethane
Topical non-steroidal agents
• Doxepin
• Hydroactive colloid gel
Laser Therapy
• Photobiomodulation therapy or low-level laser therapy
Topical corticosteroids
• Unknown steroids


70%
70%

67%
67%

15%

30%

Managing side effects associated with targeted therapies

Advances in our understanding of the mechanisms underlying cancer development have created the opportunity for new therapeutic approaches, termed ‘targeted therapies’, that selectively interfere with molecules or pathways involved in tumour growth and progression33.

Many targeted therapies work through inactivation of growth factors and their receptors that control specific functions in cancer cells33. Small-molecule inhibitors and monoclonal antibodies are the two major approaches of targeted therapy. Monoclonal antibodies target specific antigens on the extracellular surface (Figure 2), while small molecules penetrate the cell membrane to interact with intracellular proteins and enzymes (Table 4)34.

Figure 2. Monoclonal antibodies used in various cancers35. ADCC, antibody-dependent cellular cytotoxicity; APC, antigen-presenting cell; CAR-T, chimeric antigen receptor T cell; CD, cluster of differentiation; CDC, complement-dependent cytotoxicity; CTLA-4, cytotoxic T-lymphocyte associated protein 4; Fc, fragment crystallisable; HER2, human epidermal growth factor receptor 2; mAb, monoclonal antibody; MHC, major histocompatibility complex; NK, natural killer; PD-L1, programmed death-ligand 1; scFv, single-chain variable fragment; TCR, T cell receptor; VEGF, vascular endothelial growth factor.

Table 4. Small-molecule inhibitors used in various cancers34,36.

BCR-ABL, breakpoint cluster region and Abelson murine leukaemia viral oncogene homologue; CDK4/6, cyclin-dependent kinases 4 and 6; c-Kit, KIT proto-oncogene receptor tyrosine kinase; mTOR, mammalian target of rapamycin; PARP, poly (ADP-ribose) polymerase; PDGFR, platelet-derived growth factor receptors; RET, rearranged during transfection; VEGFR, vascular endothelial growth factor receptor.
Approved small-molecule inhibitors
Generic drug name Target
Imatinib BCR-ABL
Olaparib PARP
Abemaciclib, palbociclib, ribociclib CDK4/6
Selpercatinib, pralsetinib RET
Gefitinib, erlotinib EGFR
Sorafenib VEGF
Tamoxifen Oestrogen receptor
Dasatinib BCR-ABL, Sre
Sunitinib c-Kit, PDGFR, VEGFR
Temsirolimus mTOR
Lapatinib HER2
Bortezomib Proteasome 26s

Targeted therapies have fewer reported toxicities and better tolerance when compared with chemotherapy37,38. However, many targeted drugs require an extended or indefinite treatment period, which can cause long-term side effects that can be distressing for many patients. Although the incidence of side effects is lower because of target specificity, adverse effects, such as hypertension, and dermatological toxicities due to off-target drug effects are frequently observed 33,37-39.

Management of hypertension

Management of hypertension involves antihypertensive therapy, aiming to achieve a blood pressure (BP) of <130/80 mmHg39. Discontinuation of treatment is recommended if systolic BP is >180 mmHg or diastolic BP >110 mmHg39,40. First-line antihypertensive agents include angiotensin receptor blockers or angiotensin-converting enzyme (ACE) inhibitors, dihydropyridine calcium channel blockers, or thiazide or thiazide-like diuretics39. b-blockers should be reserved for individuals with a specific indication for their use, or those who did not achieve treatment success with, or have contraindications to, first-line antihypertensive agents39.

Management of dermatological side effects

Accurate grading is a critical component to determine the appropriate intervention and management response.

The most widely used grading system is the Common Terminology Criteria for Adverse Events (CTCAE), which takes into consideration the degree to which activities of daily living may be affected37,41

In CTCAE, an adverse event is defined as any abnormal clinical finding temporally associated with the use of a cancer therapy. These criteria are used for the management of treatment administration and dosing, and to provide standardisation and consistency in the definition of treatment-related toxicity37,41.

Maculopapular rash (morbilliform eruption)

Recommended treatment41:

  • Topical or oral corticosteroids
  • Antihistamines
  • Careful monitoring to prevent progression to Stevens–Johnson syndrome/toxic epidermal necrolysis (SJS/TEN)

Papulopustular (acneiform) rash

Recommended treatment includes topical corticosteroids and an oral tetracycline antibiotic (doxycycline 100 mg two times a day, minocycline 50 mg two times a day, or oxytetracycline 500 mg two times a day) for at least 6 weeks. For more severe cases, a short course of systemic corticosteroids (e.g., prednisone 0.5–1 mg/kg body weight for 7 days with a weaning dose over 4–6 weeks) can be considered37.

Xerosis

Management is done conservatively by educating the patient to minimise showering, use tepid water and avoid soaps37,38. If eczema is present, a 1–2-week course of corticosteroids is recommended. To decrease pruritis, oral antihistamines gabapentin and pregabalin can be used37.

Hand–foot skin reaction

Management of SJS/TEN using intravenous immunoglobulin (IVIg) is recommended41. For SJS, a total dose of 2 g/kg IVIg is often considered41. IVIg should be administered as soon as possible after confirming the diagnosis of TEN at a recommended total dose of 3 g/kg41.

Managing side effects of immunotherapies

The widespread use of immunotherapy has revolutionised the treatment of various cancer types. However, manipulation of the immune system to induce an anti-tumour response is associated with a unique set of immune-related side effects that differ from chemotherapy and targeted therapy toxicities, given their immune-based origin42,43. Several different classes of immunotherapies have been approved and are routinely used in cancer treatment (Table 5)44-46.

BCMA, B-cell maturation antigen; CAR, chimeric antigen receptor; CD19, cluster of differentiation 19; CTLA-4, cytotoxic T-lymphocyte associated protein 4; ISG, interferon-stimulated gene; JAK–STAT, Janus kinase-signal transducer and activator of transcription; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1.
Class Drug Target
Checkpoint inhibitors Pembrolizumab
Ipilimumab
Nivolumab
Atezolizumab
CTLA-4, PD-1, PD-L1
Cytokines Interferon
Aldesleukin
Jak-STAT, NF-kB, ISGs
CAR T cell Tisagenlecleucel
Axicabtagene ciloleucel
Brexucabtagene autoleucel
CD19, BCMA

Management of immune thrombocytopenia

Severity of immune thrombocytopenia induced by immunotherapy is graded according to platelet count, with management based on the grade of thrombocytopenia (Table 6)47.

Table 6. Incidence and management of immune thrombocytopenia according to grade of severity47,48. Proportion of treated patients affected based on patients with cancer (N=1,038) treated with immune checkpoint inhibitors. IVIg, intravenous immunoglobulin.

Grading Platelet count Proportion of treated patients affected Management
G1 ≥75 to <100/mL 32% Continue treatment with close monitoring
G2 ≥50 to <75/mL 6% Hold treatment until resolved. If not resolved, interrupt treatment until reversion to G1. Administer oral prednisone 1 mg/kg per day for 4 weeks followed by a taper over 4–6 weeks to lowest effective dose. IVIg may be used in conjunction with corticosteroids
G3 ≥25 to <50/mL 9% Dexamethasone 40 mg daily for 4 days may be considered as an alternative to prednisone.
G4 <25/mL

Management of cardiovascular side effects

A range of cardiovascular (CV) side effects have been reported after treatment with immunotherapy, including myocarditis, pericarditis, arrhythmias, cardiomyopathy, and vasculitis49,50.

Cardiovascular complications have been reported with all immune checkpoint inhibitors and, although rare, are associated with high mortality rates47,51

For patients who develop new CV symptoms after onset of treatment, a full workup should be conducted and may include47:

  • Electrocardiogram
  • Troponin and creatine phosphokinase
  • B-type natriuretic peptide (BNP)
  • Echocardiogram
  • Chest X-ray
  • Stress test
  • Cardiac catheterisation
  • Cardiac magnetic resonance imaging

If myocarditis, pericarditis, arrhythmias, impaired ventricular function with heart failure, or vasculitis is suspected, therapy should be withheld47. For symptomatic patients, high-dose corticosteroids (prednisone 1–2 mg/kg per day) should be initiated. In patients without an immediate response to high-dose corticosteroids, consider methylprednisolone 1 g/day and either mycophenolate, infliximab, or antithymocyte globulin50,51. Abatacept or alemtuzumab as additional immunosuppression should be considered in life-threatening cases47.

Management of endocrine side effects

The most common endocrine side effects associated with immunotherapy include hypothyroidism and adrenal insufficiency43,47.

Hypothyroidism

Hypothyroidism is more common than hyperthyroidism and can be detected by routine blood tests for thyroid-stimulating hormone (TSH) and free thyroxine (FT4)43,47. Once detected, routine monitoring should be done at baseline and before every infusion for 4–6 weeks while receiving treatment. Thyroid hormone supplementation can be initiated for TSH levels above 10 mIU/L47.

Adrenal insufficiency

Adrenal insufficiency is associated with inhibitors of the immune checkpoint programmed cell death protein 1 (PD-1) and its ligand PD-L143. Management depends on the clinical severity. Patients with mild signs and symptoms can be started on hydrocortisone (up to 30 mg daily); those with moderate symptoms should receive hydrocortisone 30–50 mg daily or prednisone 20 mg daily47,50. Fludrocortisone (0.05–1 mg daily) may also be required for mild or moderate symptoms47.

Patients with suspected adrenal crisis should receive an immediate dose of up to 100 mg intravenous hydrocortisone plus fluid resuscitation with 2 L saline47

Once clinically stable, maintenance oral therapy should be continued as previously described.

Management of diarrhoea

Diarrhoea can occur in up to 54% of patients treated with anti-CTLA-1 antibodies, or anti-CTLA-4 antibody and PD-1 combinations47. Management of diarrhoea depends on severity, but in moderate and severe cases treatment should be temporarily withheld until symptoms resolve47,50. Corticosteroids (1–2 mg/kg/day prednisone or equivalent) should be administered in moderate cases if diarrhoea persists, and in severe cases47,50.

Patient activation for managing cancer treatment toxicities

Patient activation, defined as an individual's knowledge, skill and confidence in managing their own health, has gained recognition for its positive impact on outcomes in individuals undergoing cancer treatment52. Research supports the notion that more activated patients monitor their conditions more closely, exhibit greater adherence to treatments, and show increased acceptance of recommended care, resulting in improved clinical outcomes. While this evidence spans different patient populations and various settings, studies focusing specifically on patient activation in patients with cancer are limited52.

With minimal clinical guidance, cancer patients and their families bear increasing responsibility for day-to-day management, encompassing symptom control, adherence to complex treatment regimens, and the adoption of behaviours that reduce the risk of disease recurrence53. As cancer survivorship increases, the need for long-term self-management by patients becomes crucial.

To address this gap, a pilot randomised trial was undertaken in 2023 to evaluate the feasibility, acceptability and preliminary effectiveness of SMARTCare (Self-Management and Activation to Reduce Treatment Toxicities), an intervention comprising an online self-management education programme (I-Can Manage) and five sessions of nurse-delivered telephone coaching, designed to improve patient activation53. Patients starting systemic therapy for lymphoma or colorectal or lung cancer at three centres in Ontario, Canada, were recruited to the study and randomised to the intervention group (SMARTCare) or an enhanced education control group53.

In this pilot study, patient-reported outcomes were evaluated using measures such as patient activation, symptom distress, self-efficacy and quality of life53. Data were collected at baseline and at multiple time points. Out of 90 approached patients, 62 (68.9%) participated, with an average age of 60.5 years. Most were married (77.1%), university educated (71%), had colorectal cancer (41.9%) or lymphoma (42.0%), and had advanced disease (75.8%). Attrition was higher in the intervention group (36.7% vs 25% in control). Adherence to the intervention was low, but the intervention group showed improvements in patient activation scores53.

The study concluded that early self-management education and coaching may enhance patient activation during cancer treatment. However, due to low adherence and attrition, a larger trial is needed to assess the feasibility and effectiveness of the intervention53.

References

  1. Was H, Borkowska A, Bagues A, Tu L, Liu JYH, Lu Z, et al. Mechanisms of Chemotherapy-Induced Neurotoxicity. Front Pharmacol. 2022;13:750507.
  2. Fabi A, Bhargava R, Fatigoni S, Guglielmo M, Horneber M, Roila F, et al. Cancer-related fatigue: ESMO Clinical Practice Guidelines for diagnosis and treatment. Ann Oncol. 2020;31(6):713-723.
  3. Wang TF, Zwicker JI, Ay C, Pabinger I, Falanga A, Antic D, et al. The use of direct oral anticoagulants for primary thromboprophylaxis in ambulatory cancer patients: Guidance from the SSC of the ISTH. J Thromb Haemost. 2019;17(10):1772-1778.
  4. Ortel TL, Neumann I, Ageno W, Beyth R, Clark NP, Cuker A, et al. American Society of Hematology 2020 guidelines for management of venous thromboembolism: treatment of deep vein thrombosis and pulmonary embolism. Blood Adv. 2020;4(19):4693-4738.
  5. Lyman GH, Carrier M, Ay C, Di Nisio M, Hicks LK, Khorana AA, et al. American Society of Hematology 2021 guidelines for management of venous thromboembolism: prevention and treatment in patients with cancer. Blood Adv. 2021;5(4):927-974.
  6. Falanga A, Ay C, Di Nisio M, Gerotziafas G, Jara-Palomares L, Langer F, et al. Venous thromboembolism in cancer patients: ESMO Clinical Practice Guideline. Annals of Oncology. 2023;34(5):452-467.
  7. National Institute for Health and Care Excellence. NICE guideline [NG158] Venous thromboembolic diseases: diagnosis, management and thrombophilia testing. Updated 2023.
  8. Gervaso L, Dave H, Khorana AA. Venous and arterial thromboembolism in patients with cancer: JACC: CardioOncology state-of-the-art review. JACC: CardioOncology. 2021;3(2):173-190.
  9. Key NS, Khorana AA, Kuderer NM, Bohlke K, Lee AYY, Arcelus JI, et al. Venous Thromboembolism Prophylaxis and Treatment in Patients With Cancer: ASCO Guideline Update. Journal of Clinical Oncology. 2023;41(16):3063-3071.
  10. National Comprehensive Cancer Network. NCCN Guidelines Version 3.2024 Hematopoietic Growth Factors. 2024.
  11. Soff G, Leader A, Al-Samkari H, Falanga A, Maraveyas A, Sanfilippo K, et al. Management of chemotherapy-induced thrombocytopenia: guidance from the ISTH Subcommittee on Hemostasis and Malignancy. Journal of Thrombosis and Haemostasis. 2024;22(1):53-60.
  12. Gao A, Zhang L, Zhong D. Chemotherapy-induced thrombocytopenia: literature review. Discover Oncology. 2023;14(1).
  13. Leader A, Hofstetter L, Spectre G. Challenges and Advances in Managing Thrombocytopenic Cancer Patients. J Clin Med. 2021;10(6).
  14. Kuter DJ. Treatment of chemotherapy-induced thrombocytopenia in patients with non-hematologic malignancies. Haematologica. 2022;107(6):1243-1263.
  15. Taplitz RA, Kennedy EB, Bow EJ, Crews J, Gleason C, Hawley DK, et al. Outpatient Management of Fever and Neutropenia in Adults Treated for Malignancy: American Society of Clinical Oncology and Infectious Diseases Society of America Clinical Practice Guideline Update. Journal of Clinical Oncology. 2018;36(14):1443-1453.
  16. Klastersky J, de Naurois J, Rolston K, Rapoport B, Maschmeyer G, Aapro M, et al. Management of febrile neutropaenia: ESMO Clinical Practice Guidelines. Ann Oncol. 2016;27(suppl 5):v111-v118.
  17. Taplitz RA, Kennedy EB, Bow EJ, Crews J, Gleason C, Hawley DK, et al. Antimicrobial Prophylaxis for Adult Patients With Cancer-Related Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update. Journal of Clinical Oncology. 2018;36(30):3043-3054.
  18. Cooper KL, Madan J, Whyte S, Stevenson MD, Akehurst RL. Granulocyte colony-stimulating factors for febrile neutropenia prophylaxis following chemotherapy: systematic review and meta-analysis. BMC Cancer. 2011;11:404.
  19. Kuderer NM, Dale DC, Crawford J, Lyman GH. 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.
  20. Aapro MS, Bohlius J, Cameron DA, Lago LD, Donnelly JP, Kearney N, 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. European Journal of Cancer. 2011;47(1):8-32.
  21. Zimmer AJ, Freifeld AG. Optimal Management of Neutropenic Fever in Patients With Cancer. J Oncol Pract. 2019;15(1):19-24.
  22. Klastersky J, Paesmans M, Rubenstein EB, Boyer M, Elting L, Feld R, et al. The Multinational Association for Supportive Care in Cancer risk index: A multinational scoring system for identifying low-risk febrile neutropenic cancer patients. J Clin Oncol. 2000;18(16):3038-3051.
  23. Jordan K, Chan A, Gralla RJ, Jahn F, Rapoport B, Ruhlmann CH, et al. Emetic risk classification and evaluation of the emetogenicity of antineoplastic agents—updated MASCC/ESMO consensus recommendation. Supportive Care in Cancer. 2024;32(1).
  24. Herrstedt J, Clark-Snow R, Ruhlmann CH, Molassiotis A, Olver I, Rapoport BL, et al. 2023 MASCC and ESMO guideline update for the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting. ESMO Open. 2024;9(2):102195.
  25. Hesketh PJ, Kris MG, Basch E, Bohlke K, Barbour SY, Clark-Snow RA, et al. Antiemetics: ASCO Guideline Update. J Clin Oncol. 2020;38(24):2782-2797.
  26. Peterson DE, Boers-Doets CB, Bensadoun RJ, Herrstedt J, Committee EG. Management of oral and gastrointestinal mucosal injury: ESMO Clinical Practice Guidelines for diagnosis, treatment, and follow-up. Ann Oncol. 2015;26 Suppl 5:v139-151.
  27. Elad S, Cheng KKF, Lalla RV, Yarom N, Hong C, Logan RM, et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer. 2020;126(19):4423-4431.
  28. Bossi P, Antonuzzo A, Cherny NI, Rosengarten O, Pernot S, Trippa F, et al. Diarrhoea in adult cancer patients: ESMO Clinical Practice Guidelines. Ann Oncol. 2018;29 Suppl 4:iv126-iv142.
  29. Common Terminology Criteria for Adverse Events (CTCAE). 2017.
  30. Larkin PJ, Cherny NI, La Carpia D, Guglielmo M, Ostgathe C, Scotte F, et al. Diagnosis, assessment and management of constipation in advanced cancer: ESMO Clinical Practice Guidelines. Ann Oncol. 2018;29 Suppl 4:iv111-iv125.
  31. Behroozian T, Goldshtein D, Ryan Wolf J, Van Den Hurk C, Finkelstein S, Lam H, et al. MASCC clinical practice guidelines for the prevention and management of acute radiation dermatitis: part 1) systematic review. eClinicalMedicine. 2023;58:101886.
  32. Behroozian T, Bonomo P, Patel P, Kanee L, Finkelstein S, Van Den Hurk C, et al. Multinational Association of Supportive Care in Cancer (MASCC) clinical practice guidelines for the prevention and management of acute radiation dermatitis: international Delphi consensus-based recommendations. The Lancet Oncology. 2023;24(4):e172-e185.
  33. McFarlane T, Rehman N, Wang K, Lee J, Carter C. Cutaneous toxicities of new targeted cancer therapies: must know for diagnosis, management, and patient-proxy empowerment. Ann Palliat Med. 2020;9(3):1296-1306.
  34. Ciavarella S, Milano A, Dammacco F, Silvestris F. Targeted therapies in cancer. . 2010;24(2):77-88.
  35. Arias-Pinilla GA, Modjtahedi H. Therapeutic Application of Monoclonal Antibodies in Pancreatic Cancer: Advances, Challenges and Future Opportunities. Cancers. 2021;13(8):1781.
  36. Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, et al. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Ther. 2021;6(1):201.
  37. Lacouture ME, Sibaud V, Gerber PA, van den Hurk C, Fernandez-Penas P, Santini D, et al. Prevention and management of dermatological toxicities related to anticancer agents: ESMO Clinical Practice Guidelines(). Ann Oncol. 2021;32(2):157-170.
  38. Huynh Dagher S, Blom A, Chabanol H, Funck-Brentano E. Cutaneous toxicities from targeted therapies used in oncology: Literature review of clinical presentation and management. Int J Womens Dermatol. 2021;7(5Part A):615-624.
  39. Cohen JB, Brown NJ, Brown S-A, Dent S, Van Dorst DCH, Herrmann SM, et al. Cancer Therapy–Related Hypertension: A Scientific Statement From the American Heart Association. Hypertension. 2023;80(3).
  40. Herrmann J, Lenihan D, Armenian S, Barac A, Blaes A, Cardinale D, et al. Defining cardiovascular toxicities of cancer therapies: an International Cardio-Oncology Society (IC-OS) consensus statement. European Heart Journal. 2022;43(4):280-299.
  41. Freites-Martinez A, Lacouture ME. Dermatologic Adverse Events. In: The MASCC Textbook of Cancer Supportive Care and Survivorship. Olver I, editor. Cham: Springer International Publishing; 2018. p. 597-620.
  42. Twomey JD, Zhang B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021;23(2):39.
  43. Gumusay O, Callan J, Rugo HS. Immunotherapy toxicity: identification and management. Breast Cancer Res Treat. 2022;192(1):1-17.
  44. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: from bench to bedside. Biomark Res. 2022;10(1):12.
  45. Baglini E, Salerno S, Barresi E, Marzo T, Settimo FD, Taliani S. Cancer Immunotherapy: An Overview on Small Molecules as Inhibitors of the Immune Checkpoint PD-1/PD-L1 (2015-2021). Mini Rev Med Chem. 2022;22(14):1816–1827.
  46. Barrat FJ, Crow MK, Ivashkiv LB. Interferon target-gene expression and epigenomic signatures in health and disease. Nat Immunol. 2019;20(12):1574-1583.
  47. Schneider BJ, Naidoo J, Santomasso BD, Lacchetti C, Adkins S, Anadkat M, et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. Journal of Clinical Oncology. 2021;39(36):4073-4126.
  48. Haddad TC, Zhao S, Li M, Patel SH, Johns A, Grogan M, et al. Immune checkpoint inhibitor-related thrombocytopenia: incidence, risk factors and effect on survival. Cancer Immunology, Immunotherapy. 2022;71(5):1157-1165.
  49. Chung WB, Youn JC, Youn HJ. Cardiovascular Complications of Novel Anti-Cancer Immunotherapy: Old Problems from New Agents? Korean Circ J. 2020;50(9):743-753.
  50. Haanen J, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Annals of Oncology. 2022;33(12):1217-1238.
  51. Brahmer JR, Lacchetti C, Thompson JA. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline Summary. J Oncol Pract. 2018;14(4):247-249.
  52. Newland P, Lorenz R, Oliver BJ. Patient activation in adults with chronic conditions: A systematic review. Journal of Health Psychology. 2021;26(1):103-114.
  53. Howell D, Pond GR, Bryant-Lukosius D, Powis M, McGowan PT, Makuwaza T, et al. Feasibility and Effectiveness of Self-Management Education and Coaching on Patient Activation for Managing Cancer Treatment Toxicities. Journal of the National Comprehensive Cancer Network. 2023;21(3):247-256.e248.
Welcome: