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:22nd May 2023
Published:25th Aug 2022

Managing side effects associated with chemotherapy and radiation therapy

Chemotherapy drugs are extensively used in clinic as one of the main cancer treatments and can be administered alone or as an 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 death.

Although these therapies have drastically improved the survival of patients, they are notorious for causing severe side effects that ultimately limit the amount that can be tolerated and thus treatment efficacy

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

Thrombosis

In this video, Dr Gary Lyman discusses the challenges of treating thrombosis in patients with cancer.

Venous thromboembolism (VTE) is a major complication of thrombosis. Anticoagulants are used for VTE prevention and treatment include unfractionated heparin (UFH), low molecular weight heparins (LMWHs), fondaparinux (a synthetic indirect inhibitor of activated factor Xa), vitamin K antagonists (VKAs) and more recently, direct oral anticoagulants (DOACS)2-7. Choice of anticoagulant is determined by considerations such as bleeding risk, patient preference and potential drug interactions3-7. 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 preferred3-7. Caution should be exercised when prescribing DOACS to people with gastrointestinal or genitourinary tract cancers because of an increased risk of bleeding3-7. If apixaban, rivaroxaban or LMWHs are contraindicated or unsuitable, alternatives such as dabigatran, UFH, fondaparinux or a VKA can be considered4.

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 VTE4,5. The ASH 2021 guidelines suggest indefinite anticoagulant treatment for the long-term prevention of VTE may be considered if the benefits outweigh the risks4.

Learn more about the management of cancer-associated thrombosis

Nausea and vomiting

In this video, Dr Lyman highlights the guideline recommended management strategies for nausea and vomiting caused by cancer treatments.

The risk of chemotherapy-induced nausea and vomiting (CINV) for each patient should be ascertained based on the type of treatment (Table 1) and predisposing factors such as gender, type of cancer and age. A useful tool recommended by the Multinational Association of Supportive Care in Cancer (MASCC) and the European Society for Medical Oncology (ESMO) for predicting CINV risk can be found at www.riskcinv.com.

Table 1. Emetogenic risk of anticancer treatments (Adapted)8.

Risk of emesis Treatment
High
(>90 %)
Cisplatin, streptozocin, carmustin, dacarbazin, antrazyclin/cyclophosphamide based regimen, cyclophosphamide ≥ 1500 mg/m2, mechlorethamine
Moderate
(30-90 %)
Alemtuzumab, arsentrioxide, azacitidine, bendamustine, carboplatin, clofarabine, cyclophosphamide < 1500 mg/m2, cytarabine > 1000 mg/m2, daunorubicin, doxorubicin, epirubicin, idarubicin, ifosfamide, interferon α, irirnotecan, oxaliplatin, romidepsin, temozolomide, thiotepa, trabectidin
Low
(10-30 %)
Aflibercept, asparaginase, belinostat, blinatumomab, bortezomib, brentuximab, cabazitaxel, cetuximab, cytarabine < 1000 mg/m2, dactinomycin, docetaxel, doxorubicin (liposomalpegyliert), eribulin, etopsid, 5-fluorouracil, gemcitabine, ipilimumab, methotrexate, mitomycin, nab-paclitaxel, paclitaxel, panitumumab, pemetrexed, pertuzumab, topotecan
Minimal
(<10 %)
Bevacizumab, bleomycin, buserelin, busulfan, 2- chlordeoxyadenosine, cladribine, fludarabine, fulvestrant, goserelin, leuprorelin, nivolumab, pembrolizumab, rituximab, trastuzumab, vinblastine, vincristine, vinorelbine

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 patients9,10.

High emetic risk

The American Society of Clinical Oncology (ASCO) guidelines recommend adults treated with cisplatin and other high-emetic-risk drugs be offered a 4-drug combination of a neurokinin-1 (NK1) receptor antagonist, a 5-hydroxytrytamine 3 (5-HT3) receptor antagonist (ondansetron, granisetron), dexamethasone (DEX) and olanzapine on day 1 of treatment. DEX and olanzapine should be continued on days 2–410. ESMO and MASCC guidelines recommend a single prophylactic dose of 5-HT3 + DEX + NK1 before chemotherapy for acute emesis in high-risk patients, with the addition of olanzapine in instances of delayed-onset emesis9.

For high-emetic risk radiation therapy, a 2-drug combination of a 5-HT3 receptor antagonist and DEX is recommended before each fraction and on the day after each fraction3

Moderate emetic risk

Adults treated with moderate-emetic risk anti-neoplastic treatments (excluding carboplatin) should be offered a 2-drug combination of a 5-HT3 receptor antagonist and DEX (day 1)9,10. ASCO further states that patients treated with cyclophosphamide, doxorubicin, oxaliplatin and other moderate-emetic-risk drugs known to cause delayed CINV may be offered DEX on days 2–310.

Patients receiving moderate-emetic-risk radiation therapy should be offered a 5-HT3 receptor antagonist before each fraction, with or without DEX, for the first 5 fractions9.

Low emetic risk

Patients treated with low-emetic-risk treatments should be offered a single dose of a 5-HT3 receptor antagonist or a single 8-mg dose of DEX before treatment9,10.

Patients undergoing low-emetic-risk radiation therapy to the brain should be offered breakthrough DEX therapy. Patients who are treated with radiation therapy to the head and neck, thorax, or pelvis should be offered breakthrough therapy with a 5-HT3 receptor antagonist, DEX, or a dopamine-receptor antagonist3.

Diarrhoea

Diarrhoea is a common side effect of various neoplastic treatments and can be classified as uncomplicated or complicated11.

To ascertain the severity of diarrhoea, it should first be graded using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE)12

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)11. Skin barrier creams should be used to prevent skin irritation caused by increased faecal contact11.

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 evaluation11.

Complicated diarrhoea

Management of complicated diarrhoea involves 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 work-up investigating the presence of blood, Clostridium difficile, Salmonella, Escherichia coli, Campylobacter and infectious colitis11.

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 mobility13.

Management with oral laxatives is an essential part of care planning, the two most recommended class of laxative being osmotic and stimulant laxatives13

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 cancer13.

Suppositories and enemas are the recommended first-line therapy where there is faecal impaction and/or where oral laxatives have failed to relieve constipation13.

Mucositis

Oral and gastrointestinal (GI) mucositis caused by high-dose chemotherapy and/or radiation continues to be an important clinical problem. A number of recommendations have been made for the prevention and treatment of oral mucositis as follows14 :

  • 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
  • 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

Guideline recommendations for prevention and treatment of GI mucositis include15:

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

Neutropenia

Watch Dr Lyman discuss guideline recommended management of febrile and non-febrile neutropenia following cancer treatment.

Most patients undergoing chemotherapy and radiation therapy will develop neutropenia, which is characterised by an absolute neutrophil count (ANC) of <500 cells/µL following treatment15.

Patients with neutropenia are highly susceptible to infection and require careful monitoring for fever, chills and sweat16

The most dangerous complication of neutropenia is febrile neutropenia. ASCO, ESMO and the Infectious Diseases Society of America (IDSA) recommend using the MASCC febrile neutropenia risk calculator to pre-emptively manage this condition15,17,18. A MASCC score of ≤20 indicates high risk, while a score of ≥21 indicates low risk17. The prophylactic use of antimicrobials to prevent febrile neutropenia is discouraged by ESMO, ASCO and the European Organisation for Research and Treatment of Cancer (EORTC)16.

Febrile neutropenia

Febrile neutropenia (FN) is defined 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/μL15. FN can be ef­fectively pre­vent­ed by administering granulocyte colony-stimulating factors (G-‍CSFs), but this is only suggested for pa­tients with a >20% risk of de­vel­op­ing FN and in those with se­ri­ous comor­bidi­ties and/‍or aged >60 years15. Several meta-analyses indicate that primary prophylaxis with G-CSF (filgrastim) reduces the risk of FN by at least 50%19,20.

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 including15,16:

  • 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

ESMO recommends use of a risk assessment algorithm to determine the appropriate management approach (Figure 1). In patients at high risk of FN, treatment with intravenous broad-spectrum antibiotics is recommended15,16. Oral antibacterial therapy can be commenced for some patients at low risk of FN, namely those who15,16:

  • 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

The majority of FN cases can be resolved with prompt empirical therapy, with tailored antibacterial therapy if the type of infection is known16.

SupportiveOncologyCare_Tab2_Figure2.png

Figure 2. ESMO algorithm of response assessment (Adapted16). ANC, absolute neutrophil count; IV, intravenous; MASCC, Multinational Association of Supportive Care in Cancer.

Monotherapy and combination therapy have equivalent efficacy; however, high-risk patients may benefit more from a ß-‍lactam an­tibi­ot­ic in com­bi­na­tion with an amino­gly­co­side16. 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 hours15,16.

Chemotherapy-induced thrombocytopenia (CIT)

Official guidelines have not codified a response to CIT, and there are few studies to describe the appropriate approach. While platelet transfusion remains the cornerstone of managing CIT, a synthesis of the available data on management practices is as follows21,22:

  • 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 10x109/L (<20x109/L if febrile)
  • Recombinant interleukin-11 (oprelvekin) is the only approved treatment for CIT, however its use is limited due to adverse effects
  • Thrombopoetin receptor agonists (TPO-RAs), such as romiplostim and eltrombopag, increase platelet production and have both been approved for immune thrombocytopenia
  • Antifibrinolytic agents such as ε-aminocaproic acid or tranexamic acid have been used in some thrombocytopenic cancer patients to improve haemostasis when platelet transfusions do not work

Fatigue

In this video, Dr Lyman discusses guideline recommended approaches to managing fatigue in cancer patients.

Management of cancer-related fatigue includes an initial screening for contributing factors such as sleep disturbance and anaemia23. Management interventions include exercise (a mixture of aerobic, strength and flexibility exercises), cognitive behavioural therapy and pharmacological management with psychostimulants such as methylphenidate and dexamphetamine23.

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 prevention and treatment of ARD24. 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 undertaken, therefore, to generate recommendations based on the opinions of 42 international ARD experts. Interventions were recommended if at least 75% consensus was reached25.

The Delphi consensus process recommended six interventions for the prevention of ARD, including photobiomodulation therapy and silicone film (in breast cancer patients), polyurethane film, mometasone furoate, betamethasone and olive oil. For the management of ARD, foam dressings were recommended (Table 2)25.

The study concluded that most interventions could not be recommended because there was insufficient evidence, conflicting evidence or lack of consensus to support use25. 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 2. Delphi consensus recommendations for interventions to prevent or manage acute radiation dermatitis (25). 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

Recent 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 progression26.

Many targeted therapies work through inactivation of growth factors and their receptors that control specific functions in cancer cells26. Small-molecule inhibitors and monoclonal antibodies are the two major approaches of targeted therapy. Monoclonal antibodies target specific antigens on the extracellular surface (Table 3), while small molecules penetrate the cell membrane to interact with intracellular proteins and enzymes (Table 4)27.

Table 3. Monoclonal antibodies used in various cancers27,28.

 CD, cluster of differentiate; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; VEGF, vascular endothelial growth factor 
Approved monoclonal antibodies
Generic drug name Target
Bevacizumab VEGF
Tositumomab, rituximab CD20
Cetuximab, panitumumab EGFR
Trastuzumab HER2
Alemtuzumab CD52
Gemtuzumab CD33

Table 4. Small-molecule inhibitors used in various cancers27,29.

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 chemotherapy30,31. 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 due to target specificity, adverse effects such as dermatological toxicities are frequently observed due to off-target drug effects26,30,31.

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 (ADLs) may be affected30,32

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 toxicity30,32.

Maculopapular rash (morbilliform eruption)

Recommended treatment30:

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

Papulopustular rash

In the case of a localised or non-extensive eruption, local treatment is usually sufficient and includes30:

  • topical antibiotics (erythromycin, clindamycin or metronidazole)
  • copper- and zinc-based antiseptic creams

Systemic treatment is prescribed when the lesions are extensive, profuse or poorly tolerated by the patient. Doxycycline 100–200 mg/d for 4–8 weeks is used as first-line therapy30.

Xerosis

Management is done conservatively by educating the patient to minimise showering, use tepid water and avoid soaps30,31. If eczema is present, a 1–2-week course of corticosteroids is recommended. To decrease pruritis, oral antihistamines, gabapentin and pregabalin can be used30.

Hand–foot skin reaction

Patient education on the signs and symptoms of hand–foot skin reaction (HFSR) is a vital part of prevention30. Certain lifestyle modifications including avoiding vigorous exercise, hot water and tight-fitting clothing should be communicated. To alleviate the symptoms of HFSR, emollients and moisturising creams can be used30.

Stevens–Johnson syndrome (SJS)/toxic epidermal necrolysis

Management of SJS/TEN using intravenous immunoglobulin (IVIg) is recommended30,32,33. IVIG should be administered as soon as possible after confirming the diagnosis of TEN at a recommended total dose of 3 g/kg33.

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 origin33,34. Several different classes of immunotherapies have been approved and are routinely used in cancer treatment (Table 5)36-38.

Table 5. Approved immunotherapy treatments36-38.

BCMA, B-cell maturation antigen; CTLA-4, CD19, cluster of differentiate 19; ISGs, interferon-stimulated genes; Jak-STAT, Janus kinase-signal transducer and activator of transcription; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells. 
Class Drug Target
Checkpoint inhibitors Pembrolizumab
Ipilumumab
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 thrombocytopenia induced by immunotherapy is graded according to platelet count, with management based on the grade of thrombocytopenia (Table 6)39.

 Table 6. Management of thrombocytopenia according to grade of severity39. IVIg, intravenous immunoglobulin.

Grading Management
G1: Platelet count <100/μL Continue treatment with close monitoring
G2: Platelet count <75/μL Hold treatment until resolved, administer oral
prednisone 1 mg/kg for 2–4 weeks
G3: Platelet count <50/μL
G4: Platelet count <25/μL Withhold treatment and refer to haematologist. Administer prednisone 1–2 mg/kg/day and IVIg 1 g/kg

Management of cardiovascular side effects

A range of cardiovascular (CV) side effects have been reported after treatment with immunotherapy, including myocarditis, pericarditis, arrhythmias and cardiomyopathy40.

Myocarditis is one of the most common CV side effects and generally occurs within 4 weeks of immunotherapy initiation39

For patients who develop new CV symptoms after onset of treatment, a full workup should be conducted including39:

  • electrocardiogram
  • troponin
  • B-type natriuretic peptide (BNP) or N-terminal-proBNP
  • C-reactive protein
  • viral titre
  • echocardiogram with global longitudinal strain
  • cardiac magnetic resonance imaging

If myocarditis is suspected, therapy should be withheld and high-dose corticosteroids (methylprednisolone 1000 mg/day followed by oral prednisone 1 mg/kg/day) should be initiated. Corticosteroids should be continued until resolution of symptoms and normalisation of troponin, left ventricular systolic function and conduction abnormalities39.

Hypertension

Management of hypertension involves antihypertensive therapy, aiming to achieve a blood pressure (BP) of <140/90 mmHg. Discontinuation of treatment is recommended if systolic BP is >200 mmHg or diastolic BP >100 mmHg. Antihypertensive therapy includes angiotensin-converting enzyme (ACE) inhibitors and beta-blockers39,40.

Management of endocrine side effects

The most common endocrine side effects associated with immunotherapy include hypothyroidism and adrenal insufficiency35,39.

Hypothyroidism

Hypothyroidism is more common than hyperthyroidism and can be detected by routine blood tests for thyroid-stimulating hormone (TSH) and free thyroxine (FT4)35,39. Once detected, routine monitoring should be done at baseline and before every infusion for 4–6 weeks whilst receiving treatment. Levothyroxine can be initiated for TSH levels above 10 mIU/L39.

Adrenal insufficiency

Adrenal insufficiency is associated with inhibitors of the immune checkpoint programmed cell death protein 1 (PD-1) and its ligand PD-L135. Management depends on the clinical severity. Patients with mild signs and symptoms can be started on prednisone (5–10 mg daily); those with moderate symptoms should receive prednisone 20 mg daily39.

Patients with suspected adrenal crisis should receive an immediate dose of 100 mg intravenous hydrocortisone plus fluid resuscitation with 2 L saline39

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

Management of diarrhoea

Diarrhoea occurs in roughly 20% of patients receiving anti-PD-1 therapy35. Management of diarrhoea depends on severity, but in moderate cases treatment should be temporarily withheld until symptoms resolve39. Corticosteroids (1 mg/kg/day) and loperamide can also be administered39.

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 treatment41. 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 cancer patients are limited and often restricted to specific cancer types41.

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 recurrence42.

Broader investigations, however, demonstrate associations between patient activation and attitudes towards cancer, satisfaction with care, and likelihood of undergoing cancer screenings. 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 SM education program (I-Can Manage) and five sessions of nurse-delivered telephone coaching, designed to improve patient activation42. 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 group42.

In this pilot study, patient-reported outcomes were evaluated using measures such as patient activation, symptom distress, self-efficacy and quality of life. 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 scores.

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 intervention42.

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. 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.
  3. 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.
  4. 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.
  5. Mandala M, Falanga A, Roila F, Group EGW. Management of venous thromboembolism (VTE) in cancer patients: ESMO Clinical Practice Guidelines. Ann Oncol. 2011;22 Suppl 6:vi85-92.
  6. National Institute for H, Care E. Venous thromboembolic diseases: diagnosis, management and thrombophilia testing: NICE Guideline. 2020.
  7. 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.
  8. Walsh D, Davis M, Ripamonti C, Bruera E, Davies A, Molassiotis A. 2016 Updated MASCC/ESMO consensus recommendations: Management of nausea and vomiting in advanced cancer. Support Care Cancer. 2017;25(1):333-340.
  9. Roila F, Molassiotis A, Herrstedt J, Aapro M, Gralla RJ, Bruera E, et al. 2016 MASCC and ESMO guideline update for the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting and of nausea and vomiting in advanced cancer patients. Ann Oncol. 2016;27(suppl 5):v119-v133.
  10. 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.
  11. 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.
  12. Common Terminology Criteria for Adverse Events (CTCAE). 2017. Available at: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf.
  13. 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.
  14. 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.
  15. Zimmer AJ, Freifeld AG. Optimal Management of Neutropenic Fever in Patients With Cancer. J Oncol Pract. 2019;15(1):19-24.
  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. 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.
  18. Wijeratne DT, Wright K, Gyawali B. Risk-Stratifying Treatment Strategies for Febrile Neutropenia-Tools, Tools Everywhere, and Not a Single One That Works? JCO Oncol Pract. 2021;17(11):651-654.
  19. 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.
  20. 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.
  21. Leader A, Hofstetter L, Spectre G. Challenges and Advances in Managing Thrombocytopenic Cancer Patients. J Clin Med. 2021;10(6).
  22. Kuter DJ. Treatment of chemotherapy-induced thrombocytopenia in patients with non-hematologic malignancies. Haematologica. 2022;107(6):1243-1263.
  23. 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.
  24. Behroozian T, Goldshtein D, Wolf JR, 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.
  25. 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.
  26. 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.
  27. Ciavarella S, Milano A, Dammacco F, Silvestris F. Targeted therapies in cancer. BioDrugs. 2010;24(2):77-88.
  28. Boyiadzis M, Foon KA. Approved monoclonal antibodies for cancer therapy. Expert Opin Biol Ther. 2008;8(8):1151-1158.
  29. 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.
  30. 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.
  31. 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.
  32. Freites-Martinez A, Lacouture ME. Dermatologic Adverse Events. In: Olver I, editor. The MASCC Textbook of Cancer Supportive Care and Survivorship. Cham: Springer International Publishing; 2018. p. 597-620.
  33. Hadaschik E, Eming R, French LE, Girolomoni G, Hertl M, Jolles S, et al. [European Guidelines (S1) on the use of high-dose intravenous immunoglobulin in dermatology]. Hautarzt. 2020;71(7):542-552.
  34. Twomey JD, Zhang B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021;23(2):39.
  35. Gumusay O, Callan J, Rugo HS. Immunotherapy toxicity: identification and management. Breast Cancer Res Treat. 2022;192(1):1-17.
  36. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: from bench to bedside. Biomark Res. 2022;10(1):12.
  37. 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.
  38. Barrat FJ, Crow MK, Ivashkiv LB. Interferon target-gene expression and epigenomic signatures in health and disease. Nat Immunol. 2019;20(12):1574-1583.
  39. 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.
  40. 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.
  41. 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.
  42. 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: