Secondary (acquired) immunodeficiency due to medical therapies
Secondary immunodeficiency can be caused by many different types of medical therapies, including cancer chemotherapy, radiation therapy, and other immunosuppressive or immunomodulatory medications.
Recently added
This page was added on 03 February 2025.
Updates made
This page was updated on [date-counter-updated-date]. View history of updates
Overview
- When feasible, all indicated vaccines should be given ≥4 weeks before starting immunosuppressive medications.
- Non-live vaccines may be administered (if needed) during immunosuppressive therapy, but doses may need to be repeated when the person is no longer immunocompromised.
- Alternatively, non-live vaccine doses may be temporarily delayed (if the risk of disease exposure is low) until the person is less immunocompromised, to optimise vaccine effectiveness.
- Generally, for people receiving immunosuppressive therapies, live vaccines should be administered ≥4 weeks before starting the treatment, if feasible. Exceptions are people receiving complement inhibitors, hydroxychloroquine, sulfasalazine and low doses of azathioprine, methotrexate and mercaptopurine, which live vaccines can be safely administered any time before, during or after treatment.
- At the end of a course of immunosuppressive therapy, the length of time before live vaccines can be given varies by agent. Reconstitution of the immune system often needs to be confirmed first.
- Combinations of immunosuppressive therapies may have a cumulative immunosuppressive effect. Providers should seek expert advice if they are uncertain about the severity of immunocompromise and whether it is safe for people receiving an immunosuppressive therapy to receive a vaccine.
For more details about the immunosuppressive potential of various medications and medical conditions, see:
- Table. Types of medical conditions and immunosuppressive therapy and associated levels of immunocompromise
- Table. Immunosuppressive potential of cancer and organ rejection therapies
- Table. Immunosuppressive potential of conventional (non-biological) immunosuppressive therapies
- Table. Immunosuppressive potential of small molecule targeted therapies
- Table. Immunosuppressive potential of biological therapies
- Table. Immunosuppressive potential of corticosteroids
- Table. Immunosuppressive potential of certain medical conditions
Introduction
Secondary immunodeficiency can be caused by many different types of medical therapies, including cancer chemotherapy, radiation therapy, and other immunosuppressive or immunomodulatory medications. Broad categories of immunosuppressive medical therapies include:
- conventional (non-biological) immunosuppressive therapies, chemotherapies and corticosteroids
- novel biological therapies and small molecule targeted therapies
Before and during treatment with immunosuppressive medications, it is important to consider:
- the immunosuppressive risks of the underlying disease and previous therapies
- the drug class and mechanism of action of the immunosuppressive therapy, including the therapeutic effect and the duration of effect on the immune system
- the consequences of combination therapies that can alter the extent and duration of immunocompromise (such as the use of corticosteroids at the same time as other conventional therapies or biological therapies)
- the risk of exposure to vaccine-preventable diseases and susceptibility to infection
Biological therapies
Biological therapies target specific cells or cytokines as a more precise approach to treating disease. The risks involved in the use of biological therapies depend on the type of immune cell or cytokine that is targeted.
The risks of immunosuppression differ between classes of biological therapies, and between medications within the same class. These risks are also heavily influenced by the underlying disease and by exposure to concurrent immunosuppressive therapies from other classes.1 As biological therapies are newer classes of medication, there are limited published data to inform the risks of immunosuppression and live vaccine administration.
Biological therapies include:
- anti-T-lymphocyte therapies such as basiliximab and abatacept. These therapies are associated with a small increased risk of infection with many common infectious organisms.
- anti-B-lymphocyte therapies such as those targeting CD20 (rituximab, ocrelizumab, ofatumumab, obinutuzumab) and CD19 (inebilizumab), and belimumab. The anti-CD20 and anti-CD19 agents can cause substantial B-cell depletion, resulting in a risk of serious bacterial and viral infections (including hepatitis B, hepatitis C and herpes zoster reactivation). This risk can persist for several months or years after therapy has finished.
- antibodies targeting cellular markers such as alemtuzumab, blinatumomab, daratumumab and elotuzumab. These agents vary in their extent of immune cell depletion. Alemtuzumab carries the highest risk of bacterial and other opportunistic infections (including tuberculosis and herpes zoster virus reactivation).
- tumour necrosis factor (TNF)-alpha inhibitors such as infliximab, etanercept, adalimumab, certolizumab and golimumab. TNF-alpha inhibition results in a modest increased risk of infection with common bacteria and mycobacteria, including Mycobacterium tuberculosis.
- interleukin (IL)-1 pathway inhibitors such as anakinra, canakinumab and rilonacept. Use of these agents results in small to moderate increases in infection risk that are mainly influenced by concurrent immunosuppressive agents and underlying comorbidities.
- IL-4, IL-5 and IgE inhibitors such as dupilumab, mepolizumab, reslizumab, benralizumab and omalizumab. These agents act by targeting pathways that drive atopy (allergy), and are unlikely to cause overall immunosuppression.
- IL-6 inhibitors such as tocilizumab and sarilumab. These agents are associated with a modest increase in common bacterial and viral infections, as well as invasive fungal infections.
- IL-17 inhibitors such as secukinumab and ixekivumab. These agents reduce the release of proinflammatory cytokines and chemokines. They have been associated with a low risk of infections (predominantly yeast infections).
- IL-12/IL-23 pathway inhibitors such as ustekinumab, risankizumab, tildrakizumab and guselkumab. These agents target the IL-12 and IL-23 pathway to protect against psoriasis, inflammatory bowel disease and ankylosing spondylitis. They are associated with a modest risk of upper respiratory tract infections, possibly reactivation of tuberculosis and chronic hepatitis B infection.
- integrin inhibitors such as natalizumab and vedolizumab. These agents block the action of integrin on the surface of immune cells and endothelial cells, which inhibits the interactions between leukocytes and intestinal blood vessels. They are associated with a small risk of herpes simplex infection and herpes zoster reactivation, and upper respiratory tract infections. Natalizumab is also associated with a small risk of the infectious complication of progressive multifocal leukoencephalopathy. Vedolizumab does not have infection risks, as it is a gut-specific integrin inhibitor
- immune checkpoint inhibitors used as a treatment for cancer include CTLA-4 inhibitors (ipilimumab) and PD-1/PD-L1 inhibitors (nivolumab or pembrolizumab). These agents are directed in their anti-cancer activity, and do not appear to be associated with an increased risk of infection2,3 or vaccine-related complications.4
- complement inhibitors such as eculizumab and ravulizumab. These agents impair the immune response to Neisseria species, as well as other encapsulated bacteria.
Some biological therapies have a mechanism of action that does not result in immunosuppression. Examples are monoclonal antibodies used for treating osteoporosis that target the calcitonin gene-related peptide (CGRP) pathway or inhibit the receptor activator of nuclear factor kappa Β (RANK) ligand.
For more details, see Table. Immunosuppressive potential of biological therapies.
Small molecule targeted therapies
Small molecule targeted therapies work by entering cells and acting on secondary messenger systems. Different types of therapies have different immunosuppressive effects, but they are generally associated with a moderately increased risk of infection.
Small molecule targeted therapies include:
- anaplastic lymphoma kinase (ALK) inhibitors, such as crizotinib, ceritinib, alectinib and brigatinib
- BCR-ABL tyrosine kinase inhibitors (such as imatinib, dasatinib, nilotinib, ponatinib and bosutinib)
- Bruton’s tyrosine kinase inhibitors (such as ibrutinib and acalabrutinib)
- cyclin-dependent kinase (CDK) inhibitors (such as palbociclib, ribociclib and abemaciclib)
- Janus kinase (JAK) inhibitors (such as tofacitinib, baricitinib and ruxolitinib)
- phosphoinositide 3-kinase (PI3K) inhibitors (such as idelalisib)
For more details, see Table. Immunosuppressive potential of small molecule targeted therapies.
Principles of non-live vaccine administration for people with secondary immunodeficiency due to medical therapies
People with secondary immunodeficiency due to medical therapies can safely receive non-live vaccines, including seasonal influenza and COVID-19 vaccines.
An exception is Q fever vaccine. At present, this vaccine is contraindicated because there are no published data on its efficacy and safety in people who are immunocompromised (see Q fever).
When feasible, people who are immunocompromised due to medical therapies should receive all age-recommended vaccines ≥4 weeks before starting immunosuppressive therapies.
However, they may receive routine non-live vaccines during immunosuppressive therapy if needed. Doses may need to be repeated when the person is no longer immunocompromised, unless an antibody response can be demonstrated using a known correlate of protection.
Some people who are immunocompromised due to medical therapies may need altered primary vaccination schedules or additional doses of some routine non-live vaccines to optimise disease protection. See Table. Recommendations for non-live vaccine administration in people who are immunocompromised due to immunosuppressive therapies.
Vaccine | Recommendation | Comments |
---|---|---|
COVID-19 |
|
Additional doses may be needed based on age, degree of immunocompromise and presence of other risk factors for severe illness from COVID-19. |
Diphtheria-tetanus-pertussis- and polio-containing vaccines a,b | Complete routine primary schedule or catch up for missed doses before starting the immunosuppressive therapies. | – |
Haemophilus influenzae type ba,b | Complete routine primary schedule or catch up for missed doses before starting the immunosuppressive therapies. | – |
Hepatitis B a,b | 1 booster dose for seronegative people with specific at risk conditions | – |
Human papillomavirus | Aged ≥9 years: a 3-dose primary schedule | Vaccination may also be considered in younger people who are immunocompromised if there is risk of exposure to HPV |
Influenza | 1 dose annually |
|
Japanese encephalitis (inactivated) | Use if indicated | See also Vaccination for international travellers. |
Meningococcal |
For people with acquired complement deficiency due to complement inhibitor:
|
The number of doses depends on the vaccine brand and the person’s age when they start the vaccine course. See Table. Recommendations for MenACWY vaccine for people with a specified medical condition that increases their risk of invasive meningococcal disease and Table. Recommendations for MenB vaccine for people with a specified medical condition that increases their risk of invasive meningococcal disease |
Mpox (live, non-replicating) | Use if indicated or seek specialist advice on individual risks and benefits of vaccination | – |
Pneumococcal(conjugate and polysaccharide vaccines) |
Routine age-appropriate primary course and following additional doses are recommended:
|
|
Rabies | Use if indicate | See also Travellers who are immunocompromised. |
Immunisation against respiratory syncytial virusa | Adults ≥60 years: 1 primary dose of RSV vaccine At-risk neonates and infants aged <24 months: 1 primary dose of RSV-specific monoclonal antibody | – |
Zoster (herpes zoster, recombinant) | Aged ≥18 years if immunocompromised: a 2-dose primary schedule | – |
Acronyms used:
Footnotes: a Recommended but not funded under the National Immunisation Program |
Principles of live vaccine administration for people with secondary immunodeficiency due to medical therapies
Live vaccines are generally contraindicated during immunosuppressive therapy due to the risk of disseminated disease. Exceptions are for people receiving the following as monotherapy:
- complement inhibitors
- single immune checkpoint inhibitor used as anti-neoplastic agents (although data is limited in this population) – see Immune checkpoint inhibitors
- low doses of certain conventional immunosuppressive therapies — see Conventional (non-biological) immunosuppressive therapies
- certain targeted biological or small molecular therapies — see Biological therapies and Small molecule targeted therapies
People receiving other medications should receive any recommended live vaccines ≥4 weeks before starting the immunosuppressive treatment.
After therapy, the length of time before live vaccines can be given varies by agent. Immune reconstitution should be confirmed before giving live vaccines.
For therapies that are not included in this section or related tables, seek advice from the treating specialist about infection risk and safety of live vaccines.
See Table. Recommended timing of live vaccine administration for people receiving immunosuppressive therapies. If combinations of immunosuppressive therapies have been prescribed, seek specialist advice.
Category of therapy | Type of therapy and examples (not an exhaustive list) | Timing of live vaccine administration |
---|---|---|
Immunosuppressive therapies for malignancies or organ transplant | Single immune checkpoint inhibitor (e.g. ipilimumab, nivolumab) | Any time before, during or after treatment (beware of a theoretical risk of immune-mediated adverse events). |
Conventional chemotherapy or radiotherapy |
|
|
Conventional anti-rejection agents used after solid organ transplant |
|
|
Conventional immunosuppressive therapies for autoimmune, inflammatory or rheumatic diseases |
|
Any time before, during or after treatment |
|
|
|
|
|
|
Biological therapies |
|
Any time before, during or after treatment |
|
|
|
|
|
|
B-cell inhibitors (e.g. rituximab) |
|
|
Small molecule targeted therapiesb |
|
Any time before, during or after treatment |
|
|
|
Acronyms used:
Footnotes: a People taking azathioprine (≤3 mg/kg/day), 6-mercaptopurine (≤1.5 mg/kg/day), methotrexate (≤25 mg/week) can receive live vaccines on advice from a specialist and after a risk assessment. |
Corticosteroid therapy: recommendations for vaccination
Overview
People receiving low-dose corticosteroids (<2 mg/kg/day for children, or <20 mg/day for adults, prednisolone-equivalent dosing) over a short time (<14 days) can safely receive non-live and live vaccines.
People receiving corticosteroids at a higher dose (≥2 mg/kg/day for children, or ≥20 mg/day for adults, prednisolone-equivalent dosing) or over a longer time (≥14 days) can safely receive non-live and live vaccines, either:
- 4 weeks before starting high-dose corticosteroids, or
- ≥4 weeks after treatment ends, or
- during a period when corticosteroids are weaned (to <20 mg/day for adults, or <2 mg/kg/day for children, prednisolone-equivalent dosing) for at least 1 month
Specialist guidance should be sought on administration of live vaccines in people taking prolonged or high-dose corticosteroids alongside other immunosuppressive therapies.
Introduction
Corticosteroids have anti-inflammatory and immunosuppressive effects that are reversible when the medication is discontinued. Several topical, inhaled and systemic corticosteroids are available in Australia. These agents differ with respect to their potency and duration of action, which determines the corticosteroid’s efficacy and therapeutic use.
The level of immunosuppression caused by corticosteroids is affected by the specific corticosteroid prescribed, the dose, therapy duration and route of administration. The minimum dose of corticosteroid therapy that causes immunosuppression is not well defined.
In this Handbook, corticosteroid doses are defined against prednisolone as a standard.5-7 Prednisolone is one of the most widely used systemic corticosteroids, and is therefore helpful as a reference agent for people taking other systemic corticosteroids. See Table. Prednisolone-equivalent dosing, duration of action and anti-inflammatory activity for various systemic corticosteroid preparations.
Glucocorticoids (by duration of action) | Agent | Anti-inflammatory activity relative to prednisolone activitya | Approximate equivalent dose (mg) relative to prednisolone | Duration of action (hours) |
---|---|---|---|---|
Short-acting glucocorticoids | Hydrocortisone | 0.25 | 4 | 8–12 |
Cortisone acetate | 0.2 | 5 | 8–12 | |
Intermediate-acting glucocorticoids | Prednisone | 1 | 1 | 12–36 |
Prednisolone | 1 | 1 | 12–36 | |
Methylprednisolone | 1.25 | 0.8 | 12–36 | |
Triamcinolone | 1.25 | 0.8 | 12–36 | |
Long-acting glucocorticoids | Dexamethasone | 7.5 | 0.15 | 36–72 |
Betamethasone | 7.5 | 0.15 | 36–72 | |
Mineralocorticoids | Fludrocortisone | Although fludrocortisone at clinically relevant doses is not regarded as an anti-inflammatory agent, its anti-inflammatory potency is 2.5 to 3.75 times that of prednisolone | na | 12–36 |
Acronyms used:
Footnotes: a Equivalent anti-inflammatory dose shown is for oral or intravenous administration. Source: Table adapted from Furst and Saag, 2023, Determinants of glucocorticoid dosing.7 |
‘High-dose’ corticosteroids
‘High-dose’ corticosteroids
International consensus considers ‘high-dose’ corticosteroid therapy for adults to be at least 20 mg of prednisolone (or equivalent) for more than 2–4 weeks. These dosing thresholds are consistent across various global advisory groups.8-12
This definition is mainly based on expert consensus. There is a lack of rigorous scientific evaluation to clearly understand how steroid dosing impacts vaccine effectiveness and safety. The immunosuppressive risks of corticosteroids also vary for different infections.13-15
This definition of ‘high-dose’ corticosteroids for immunosuppressive effects differs from the definition of high-dose corticosteroids for treatment purposes (usually defined as 30–100 mg/day, which is the dose that results in almost complete cytosolic receptor saturation).16
People receiving ‘high-dose’ corticosteroids may be at risk of:
- reactivation of vaccine-preventable diseases17-19
- diseases caused by other pathogens20,21
- adverse events after receiving live vaccines
Vaccine safety and efficacy for people prescribed corticosteroids
Generally, there are no vaccine efficacy or safety concerns for people receiving low-dose corticosteroids (<20 mg/day for adults, or <2 mg/kg/day for children) over a short course (<14 days).22-24 These people can safely receive non-live and live vaccines.
While there is evidence suggesting that long-term, high-dose corticosteroids may impair vaccine-based immunogenicity, the effect on clinical efficacy or effectiveness of vaccines is limited.25,26 Additional booster doses may be advised to optimise vaccine efficacy, especially in high-risk children.
Individuals taking high doses of prednisolone (≥20 mg/day in adults, or >2mg/kg/day in children) for at least 4 weeks may have a higher risk of complications following live vaccine administration, due to the impact on B-cell function and immunoglobulin levels.27
Inhaled, topical and physiologically dosed corticosteroids
Generally, non-systemic corticosteroids (such as inhaled, topical use or locally injected preparations) do not cause substantial systemic immunosuppression.
Similarly, long-term physiologically dosed corticosteroid replacements (such as for people with adrenal insufficiency, defined as 3–5 mg/day for adults, or 8–10 mg/m2/day for children, prednisolone-equivalent dosing) do not cause significant immunosuppression.28
People taking these treatments can safely receive all non-live and live vaccines.
High-dose oral and intravenous injection of corticosteroids, including pulse therapy
People who receive high-dose corticosteroids (≥20 mg/day for adults, or ≥2 mg/kg/day for children, prednisolone-equivalent dosing) for at least 14 days should receive live vaccines 4 weeks before starting therapy, or at least 4 weeks after treatment ends.
Alternatively, corticosteroids should be weaned to less than 20 mg/day for at least 1 month to enable the person to safely receive live vaccines.
A single dose of dexamethasone to manage an acute respiratory illness in children is not associated with a decrease in endogenous corticosteroid levels or immunosuppression.29 No minimum interval is required between short-term dexamethasone therapy and a live vaccine, as long as the person is not acutely unwell. See Table. Recommended timing of live vaccine administration for people receiving corticosteroids.
Administration route and dose type | Corticosteroid agent and dose regimens | Administration of live vaccines |
---|---|---|
Local administration, any dose |
Any agent:
|
Any time before, during or after treatment |
Oral administration, any dose | Budesonide | Any time before, during or after treatment |
Fluticasone | ||
Single dose of dexamethasonea | ||
Physiological maintenance doses (replacement therapy) | Hydrocortisone, prednisolone or fludrocortisoneb | Any time before, during or after treatment |
Non-physiological (maintenance) doses, long-term treatment | Hydrocortisone |
|
Oral administration, prednisolone-equivalent dosec for infants and children <10 kg | <1 mg/kg/day for <28 days | Any time before, during or after treatment |
<2 mg/kg/day for <14 days | ||
<2 mg/kg/day for ≥14 days |
|
|
≥2 mg/kg/day for <14 days | ||
≥2 mg/kg/day for ≥14 days |
|
|
Oral administration, prednisolone-equivalent dosec for children aged <16 years, weight >10 kg | <10 mg/day for <28 days | Any time before, during or after treatment |
<20 mg/day for <14 days | ||
<20 mg/day for ≥14 days |
|
|
≥20 mg/day for <14 days | ||
≥20 mg/day for ≥14 days |
|
|
Oral administration, prednisolone-equivalent dosec for children aged ≥16 years and adults | <20 mg/day, any duration | Any time before, during or after treatment |
≥20 mg/day for <14 days |
|
|
≥20 mg/day for ≥14 days |
|
|
Footnotes: a A single dose of dexamethasone to manage an acute respiratory illness in children is not associated with immunosuppression. These children can receive live vaccines before or after the dose. b Physiological maintenance doses include a daily dose of 15–25 mg hydrocortisone or equivalent in people with adrenal insufficiency (such as 3–5 mg/day for adults, or 8–10 mg/m2 body surface area/day for children, prednisolone-equivalent dosing). Physiological maintenance doses of fludrocortisone (0.05–0.2 mg/day) may be prescribed for mineralocorticoid replacement levels in adrenocortical insufficiency. c Prednisolone-equivalent dose of other glucocorticoids should be used to assess a person’s suitability to receive live viral vaccines. See Table. Prednisolone-equivalent dosing, duration of action and anti-inflammatory activity for various systemic corticosteroid preparations. |
Conventional (non-biological) immunosuppressive therapies: recommendations for vaccination
Conventional immunosuppressive therapies act broadly to reduce the inflammatory responses associated with immune-mediated inflammatory disease. These include conventional non-biological medications used for treating rheumatic diseases (also known as disease-modifying anti-rheumatic drugs), autoimmune inflammatory diseases, cancer or organ rejections, such as:
- azathioprine
- calcineurin inhibitors (cyclophosphamide, tacrolimus, cyclosporin)
- hydroxychloroquine
- leflunomide
- methotrexate
- mercaptopurine
- mycophenolate
- sirolimus
- sulfasalazine
Each conventional immunosuppressive therapy has a unique mechanism of action in the inflammatory cascade and suppresses the immune system to varying degrees. See Table. Immunosuppressive potential of conventional (non-biological) immunosuppressive therapies.
Non-live vaccines
People taking conventional immunosuppressive therapy in the short or medium term can safely receive non-live vaccines. However, the immune response may be suboptimal. If non-live vaccines are received within 4 weeks of starting treatment or during treatment, these doses should be repeated after treatment and when immune competence is restored.
Live vaccines
Live vaccines can be safely administered to people receiving:
- hydroxychloroquine and sulfasalazine
- low-dose methotrexate (≤25 mg/week), azathioprine (≤3 mg/kg/day) or mercaptopurine (≤1.5 mg/kg/day)
People prescribed other conventional immunosuppressive therapy should receive live vaccines ≥4 weeks before starting, or ≥3 months after treatment ends.30 Exceptions are people receiving mTOR (mammalian target of rapamycin) inhibitors (when used for treatment of rheumatic diseases, not for preventing organ rejection) and leflunomide who can receive live vaccines ≥4 weeks after treatment ends. See Table. Recommended timing of live vaccine administration for people receiving immunosuppressive therapies.
For people receiving leflunomide, there are minimal data on the safety of live vaccines.31 Leflunomide has a low risk of immunosuppression based on its mechanism of action. The use of live vaccines during therapy may also be considered after an individual risk–benefit assessment.
Biological therapies: recommendations for vaccination
Biological therapies do not cause generalised immunosuppression like conventional immunosuppressive therapies (such as disease-modifying anti-rheumatic drugs and corticosteroids). However, biological therapies can affect the immune system, which may compromise host defences and increase the risk of serious infections.
Non-live vaccines
People receiving biological therapy in the short or medium term can safely receive non-live vaccines. However, the immune response may be suboptimal. If non-live vaccines are received within 4 weeks of starting treatment or during treatment, these doses should be repeated after treatment and when immune competence is restored.
Live vaccines
People taking complement inhibitors, anti-IL-5, anti-IgE, anti-RANK ligand or anti-CGRP therapies can receive live vaccines at any time before, during or after the treatment.
People taking other classes of biological therapies should receive recommended live vaccines ≥4 weeks before starting treatment, where possible.
After therapy, the length of time before live vaccines can be given varies by therapy. See:
- Table. Recommended timing of live vaccine administration for people receiving immunosuppressive therapies
- Table. Immunosuppressive potential of biological therapies
People receiving anti-B-cell antibodies directed against CD20 (such as rituximab) or CD52 (such as alemtuzumab) should not receive live vaccines until ≥6 months after therapy ends. This is because these agents cause substantial B-cell depletion that lasts for several months. This timing may also be affected by any blood products the person has received, including intravenous immunoglobulin (see Vaccination for people who have recently received normal human immunoglobulin and other blood products).
Infants exposed to TNF-alpha inhibitors in the 2nd or 3rd trimester in utero can receive live vaccines according to routine schedules.32 However, infants exposed to anti-CD20 B-cell antibodies (such as rituximab) should not receive live vaccines until >6 months of age (see Infants exposed to immunosuppressive therapy in utero or through breastmilk).30
Combinations of immunosuppressive therapies may have a cumulative immunosuppressive effect. Providers should seek expert advice if they are uncertain about the severity of immunocompromise and whether it is safe for people receiving biological therapies to receive a vaccine.
Immune checkpoint inhibitors (as a cancer treatment): recommendations for vaccination
Immune checkpoint inhibitors are immunomodulatory antibodies that block checkpoint proteins from binding with their partner proteins. This stimulates the immune system to attack cancer cells. These drugs include:
- CTLA-4 inhibitors such as ipilimumab and tremelimumab
- PD-1 and PD-L1 inhibitors such as nivolumab, pembrolizumab and atezolizumab
Immune checkpoint inhibitors are a comparatively recent drug class with limited available evidence so far. The risk of immunosuppression appears to be low when they are used as monotherapy.33
Non-live vaccines
People who are being treated with a single immune checkpoint inhibitor can receive non-live vaccines.
Some studies suggest that people receiving multiple immune checkpoint inhibitors may have a higher risk of adverse events following immunisation with influenza vaccine34,35 than those on a single checkpoint inhibitor.36
Live vaccines
There are no conclusive data on the safety or administering live vaccines to people receiving immune checkpoint inhibitors as a treatment for cancer. Although these therapies are not immunosuppressive given their directed mechanism of action when used as a treatment for cancer, there is a theoretical concern about a potential risk of immune-related adverse events.4
Complement inhibitors: recommendations for vaccination
Complement inhibitors (including eculizumab and ravulizumab) may be prescribed to treat atypical haemolytic uraemic syndrome, paroxysmal nocturnal haemoglobinuria and myasthenia gravis. These agents target the membrane attack complex of the complement cascade, without affecting any other immune system cells.
People receiving complement inhibitors can receive all recommended live and non-live vaccines according to routine schedules.
During therapy, people receiving complement inhibitors are at high risk of infection with Neisseria meningitidis and other Neisseria species, as well as other encapsulated bacteria. This means that:
- At least 2 weeks before starting complement inhibitor therapy, people should have received a full primary course of MenACWY (meningococcal ACWY) and MenB (meningococcal B) vaccines, and routinely recommended primary courses of pneumococcal and Hib (Haemophilus influenzae type b) vaccines,37,38 if possible.
- During therapy, people should receive booster doses of MenACWY and MenB vaccines. The number of doses for primary vaccine courses and booster timing depends on the vaccine brand and age. See recommendations for MenACWY and MenB vaccines for people at risk of invasive meningococcal disease.
- Even if they are fully vaccinated, people receiving complement inhibitor therapy remain at higher risk of infections, especially invasive meningococcal disease.39 Providers should advise patients of the ongoing risk of infection and consider antibiotic prophylaxis, especially if treatment starts before vaccination schedules are complete.
Small molecule targeted therapies: recommendations for vaccination
Small molecule targeted therapies target intracellular pathways to decrease the signalling of several cytokine and growth factor receptors. They are usually prescribed as oral medications and have a short half-life. Small molecule targeted therapies are generally associated with a moderate risk of infections.33
Non-live vaccines
People receiving small molecule targeted therapies can safely receive non-live vaccines. However, the immune response may be suboptimal due to blunted humoral and cell-mediated responses.
Live vaccines
Live vaccines are often contraindicated in people receiving small molecule targeted therapies because the underlying disease in these people is usually a malignancy. But there are some exceptions:
- JAK inhibitors are often prescribed to treat autoimmune disorders. People receiving JAK inhibitors should receive any live vaccines ≥4 week before starting therapy or ≥4 weeks after therapy ends. If therapy is likely to be long-term, doses may be withheld to enable live vaccines to be safely administered.30
- Children on BCR-ABL inhibitors have safely received live vaccines. The vaccines were well tolerated but seroconversion did not always occur.40
- ALK inhibitors and other tyrosine kinase inhibitors target neoplastic cells in non-small cell lung cancer. Live vaccines may be considered for people receiving ALK inhibitors, as no safety concerns have been reported. Consult with the treating oncologist for an individual risk–benefit assessment.
See Table. Recommended timing of live vaccine administration for people receiving immunosuppressive therapies for more details.
References
- Mathias CB, McAleer JP, Szollosi DE. Pharmacology of immunotherapeutic drugs. Cham, Switzerland: Springer; 2020.
- Fujita K, Kim YH, Kanai O, et al. Emerging concerns of infectious diseases in lung cancer patients receiving immune checkpoint inhibitor therapy. Respiratory Medicine 2019;146:66-70.
- Del Castillo M, Romero FA, Arguello E, et al. The spectrum of serious infections among patients receiving immune checkpoint blockade for the treatment of melanoma. Clinical Infectious Diseases 2016;63:1490-3.
- Spagnolo F, Boutros A, Croce E, et al. Influenza vaccination in cancer patients receiving immune checkpoint inhibitors: a systematic review. European Journal of Clinical Investigation 2021;51:e13604.
- Liu D, Ahmet A, Ward L, et al. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol 2013;9:30.
- Brunton LL, Chabner BA, Knollmann BC. Goodman and Gilman's The pharmacological basis of therapeutics. 12th ed. New York, NY: McGraw-Hill Education; 2011.
- Furst DE, Saag KG. Determinants of glucocorticoid dosing. In: Seo P, ed. UpToDate. Wolters Kluwer; 2023. (Accessed 11/1/2025). http://www.uptodate.com/contents/determinants-of-glucocorticoid-dosing?…
- Public Health Agency of Canada. Immunosuppressive therapy. In: Immunization of immunocompromised persons: Canadian immunization guide. Ottawa: Government of Canada; 2023. (Accessed 11/1/2025). https://www.canada.ca/en/public-health/services/publications/healthy-li…
- Health New Zealand. Section 4: Immunisation of special groups – 4.3 Immunocompromised individuals. In: Immunisation handbook 2024, version 5. Wellington: Ministry of Health; 2024. (Accessed 11/1/2025). https://www.tewhatuora.govt.nz/for-health-professionals/clinical-guidan…
- UK Health Security Agency. Chapter 6: Contraindications and special considerations. In: The green book. London: GOV.UK; 2017. https://www.gov.uk/government/publications/contraindications-and-specia…
- Immunization in special clinical circumstances. In: Pickering L, Baker C, Kimberlin D, Long S, eds. Red Book: Report of the Committee on Infectious Diseases. Washington, DC: American Academy of Pediatrics; 2009.
- Centers for Disease Control and Prevention (CDC). Vaccine recommendations and guidelines of the ACIP: General best practice guidelines for immunization – Altered immunocompetence. Atlanta, GA: CDC; 2023. https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/immunocompetenc…
- Park JW, Curtis JR, Lee H, et al. Risk-benefit analysis of isoniazid monotherapy to prevent tuberculosis in patients with rheumatic diseases exposed to prolonged, high-dose glucocorticoids. PLoS One 2020;15:e0244239.
- Kim MH, Choi SR, Park JK, et al. Risk of bloodstream infection in patients with systemic lupus erythematosus exposed to prolonged medium-to-high-dose glucocorticoids. Lupus 2023;32:625-32.
- Dixon WG, Abrahamowicz M, Beauchamp ME, et al. Immediate and delayed impact of oral glucocorticoid therapy on risk of serious infection in older patients with rheumatoid arthritis: a nested case-control analysis. Annals of the Rheumatic Diseases 2012;71:1128-33.
- Buttgereit F, da Silva JA, Boers M, et al. Standardised nomenclature for glucocorticoid dosages and glucocorticoid treatment regimens: current questions and tentative answers in rheumatology. Annals of the Rheumatic Diseases 2002;61:718-22.
- Wu Q, Liu Y, Wang W, et al. Incidence and prevalence of tuberculosis in systemic lupus erythematosus patients: a systematic review and meta-analysis. Frontiers in immunology 2022;13:938406.
- Pappas DA, Hooper MM, Kremer JM, et al. Herpes zoster reactivation in patients with rheumatoid arthritis: analysis of disease characteristics and disease-modifying antirheumatic drugs. Arthritis Care and Research 2015;67:1671-8.
- Cheng J, Li JB, Sun QL, Li X. Reactivation of hepatitis B virus after steroid treatment in rheumatic diseases. Journal of Rheumatology 2011;38:181-2.
- Fardet L, Genereau T, Poirot JL, et al. Severe strongyloidiasis in corticosteroid-treated patients: case series and literature review. Journal of Infection 2007;54:18-27.
- Fischer L, Gerstel PF, Poncet A, et al. Pneumococcal polysaccharide vaccination in adults undergoing immunosuppressive treatment for inflammatory diseases – a longitudinal study. Arthritis Res Ther 2015;17:151.
- Masten B, McWilliams B, Lipscomb M, et al. Immune response to hepatitis B vaccine in asthmatic children. Pediatric Pulmonology 2003;36:522-8.
- Johnson JR, Denis R, Lucas CE, et al. The effect of steroids for shock on the immune response to tetanus toxoid. American Surgeon 1987;53:389-91.
- Fairchok MP, Trementozzi DP, Carter PS, Regnery HL, Carter ER. Effect of prednisone on response to influenza virus vaccine in asthmatic children. Archives of Pediatrics and Adolescent Medicine 1998;152:1191-5.
- Crowe SR, Merrill JT, Vista ES, et al. Influenza vaccination responses in human systemic lupus erythematosus: impact of clinical and demographic features. Arthritis and Rheumatism 2011;63:2396-406.
- Abu-Shakra M, Press J, Varsano N, et al. Specific antibody response after influenza immunization in systemic lupus erythematosus. Journal of Rheumatology 2002;29:2555-7.
- Price NB, Grose C. Corticosteroids contribute to serious adverse events following live attenuated varicella vaccination and live attenuated zoster vaccination. Vaccines 2021;9:23.
- Bornstein SR, Allolio B, Arlt W, et al. Diagnosis and treatment of primary adrenal insufficiency: an Endocrine Society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism 2016;101:364-89.
- Gill N, Sirizzotti N, Johnson D, et al. Endogenous glucocorticoid response to single-dose dexamethasone for croup in children: a pharmacodynamic study. Pediatric Emergency Care 2020;36:50-6.
- Bass AR, Chakravarty E, Akl EA, et al. 2022 American College of Rheumatology guideline for vaccinations in patients with rheumatic and musculoskeletal diseases. Arthritis Care and Research 2023;75:449-64.
- Dos Reis BS, Staub FC, Koishi A, et al. Seroconversion of rheumatoid arthritis patients after yellow fever vaccination. Clinical Rheumatology 2022;41:705-8.
- Weiss B, Ben-Horin S, Lev A, et al. Immune function in newborns with in-utero exposure to anti-TNFα therapy. Front Pediatr 2022;10:935034.
- Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious complications of biological and small molecule targeted immunomodulatory therapies. Clinical Microbiology Reviews 2020;33:e00035-19.
- Laubli H, Balmelli C, Kaufmann L, et al. Influenza vaccination of cancer patients during PD-1 blockade induces serological protection but may raise the risk for immune-related adverse events. J Immunother Cancer 2018;6:40.
- Schenk EL. Clinical outcomes of patients on check point inhibitor therapy who receive routine vaccinations. Journal of Clinical Oncology 2017;35(15 Suppl):Abstract e14597.
- Wijn DH, Groeneveld GH, Vollaard AM, et al. Influenza vaccination in patients with lung cancer receiving anti-programmed death receptor 1 immunotherapy does not induce immune-related adverse events. European Journal of Cancer 2018;104:182-7.
- Heo YA. Pegcetacoplan: a review in paroxysmal nocturnal haemoglobinuria. Drugs 2022;82:1727-35.
- Therapeutic Goods Administration. Australian Public Assessment Report for pegcetacoplan. Canberra: Australian Government Department of Health; 2022. https://www.tga.gov.au/sites/default/files/2022-09/auspar-pegcetacoplan…
- McNamara LA, Topaz N, Wang X, et al. High risk for invasive meningococcal disease among patients receiving eculizumab (Soliris) despite receipt of meningococcal vaccine. MMWR. Morbidity and Mortality Weekly Report 2017;66:734-7.
- Bettoni da Cunha-Riehm C, Hildebrand V, Nathrath M, Metzler M, Suttorp M. Vaccination with live attenuated vaccines in four children with chronic myeloid leukemia while on imatinib treatment. Frontiers in immunology 2020;11:628.