Skip to main content
Annals of Oncology logoLink to Annals of Oncology
. 2018 Apr 24;29(6):1354–1365. doi: 10.1093/annonc/mdy117

Anti-infective vaccination strategies in patients with hematologic malignancies or solid tumors—Guideline of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO)

C T Rieger 1,, B Liss 2,#, S Mellinghoff 3,4,#, D Buchheidt 5, O A Cornely 3,4,6, G Egerer 7, W J Heinz 8, M Hentrich 9, G Maschmeyer 10, K Mayer 11, M Sandherr 12, G Silling 13, A Ullmann 8, M J G T Vehreschild 2, M von Lilienfeld-Toal 14, H H Wolf 15, N Lehners 7,16,#; German Society of Hematology and Medical Oncology Infectious Diseases Working Group (AGIHO)
PMCID: PMC6005139  PMID: 29688266

Abstract

Infectious complications are a significant cause of morbidity and mortality in patients with malignancies specifically when receiving anticancer treatments. Prevention of infection through vaccines is an important aspect of clinical care of cancer patients. Immunocompromising effects of the underlying disease as well as of antineoplastic therapies need to be considered when devising vaccination strategies. This guideline provides clinical recommendations on vaccine use in cancer patients including autologous stem cell transplant recipients, while allogeneic stem cell transplantation is subject of a separate guideline. The document was prepared by the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO) by reviewing currently available data and applying evidence-based medicine criteria.

Keywords: infection, anti-infective vaccination, cancer, immunosuppression, autologous stem cell transplantation


Key Message

Cancer patients are at an increased risk of infectious complications. Vaccination can be an effective preventive strategy against many infections, but type and timing of vaccinations have to be carefully chosen to allow for optimal immunization in immunocompromised patients. This review offers a comprehensive overview of vaccination strategies in different cancer populations.

Introduction

In patients with malignancies, infections significantly contribute to morbidity and mortality by delaying or impeding appropriate antineoplastic treatment. Besides prophylactic treatment, vaccination is effective in preventing infections. Patients receiving cancer treatments may experience potentially harmful long-term immunocompromising side effects and loss of previous immunizations. Such cancer survivors deserve additional care to prevent infections, but uncertainties exist concerning optimal vaccination strategies, including the choice of vaccine (passive or active, dead or life-attenuated) and time-schedules of vaccination. Therefore, the Strength of Recommendation/Quality of Evidence sometimes applies in general for the respective recommendations and not necessarily on specific settings (e.g. prime-boost-strategy for pneumococcal vaccine).

This guideline was developed to aid clinicians with decisions concerning vaccination strategies in cancer patients and recipients of autologous stem cell transplants (SCTs). These recommendations apply to the epidemiological situation in Germany, but are also applicable for regions with similar epidemiologic features.

Methods

This guideline was developed by an expert panel designated by the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO). The panel consisted of 17 specialists of internal medicine, hematology, medical oncology and/or infectious diseases led by a guideline coordinator (CTR). After definition of topics regarding specific malignant conditions and therapeutic situations, a systematic search of MEDLINE and recent scientific meetings’ databases was performed to identify relevant publications which were then thoroughly reviewed and rated. Following data extraction and assessment, preliminary recommendations were first discussed and revised by the specialist panel, then discussed and agreed upon by the AGIHO general assembly resulting in the final guideline presented here.

For grading of strength of recommendation and quality of evidence, the grading system proposed by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) was applied (Table 1) [1].

Table 1.

ESCMID grading system of strength of recommendation and quality of evidence [1]

Strength of recommendation
A Strongly support a recommendation for use
B Moderately support a recommendation for use
C Marginally support a recommendation for use
D Support a recommendation against use
Quality of evidence
I Evidence from at least one properly designed randomized, controlled trial
II* Evidence from at least one well-designed clinical trial, without randomization; from cohort- or case-control analytic studies (preferably from more than one center); from multiple time series; or from dramatic results from uncontrolled experiments
III Evidence from opinion of respected authorities, based on clinical experience, descriptive case studies, or report of expert committees
*Added index
r Meta-analysis or systematic review of randomized controlled trials
t Transferred evidence, i.e. results from different patients’ cohorts or similar immune status situation
h Comparator group is a historical control
u Uncontrolled trial
a Abstract published at an international meeting

A general limitation in vaccination studies is that success of vaccination is mostly measured by immune response to vaccination. While in some cases also the impact on incidence of infections is reported, usually no detailed information on the reduction of infection rates due to the specific sero-/genotypes against which patients had been immunized has been reported.

General recommendations

The basis of decision-making should include the knowledge of preventable diseases for which vaccination is available and feasible in immunocompromised patients. The risk of severe infection is influenced by the underlying malignancy and by the cancer-specific therapies. Therefore, vaccination recommendations for cancer patients should be based on these two major aspects.

Moreover, close family members or contact persons should be evaluated for their current vaccination status and possibly be (re-)vaccinated. Special attention is required in patients with small children, as some live-attenuated childhood vaccines may bear risks for chemotherapy recipients. For instance, Rota-virus vaccination is strongly discouraged in family members of cancer patients (DIII).

An overview of all vaccinations discussed in this review in the context of underlying malignancies and specific therapies is given in the supplementary Table S1, available at Annals of Oncology online.

Disease entity-specific vaccination strategies

Here, disease entity-specific vaccination strategies are delineated. An overview of disease entity-specific vaccination strategies is given in Table 2.

Table 2.

Disease entity-specific vaccination strategies

Tetanus Diphtheria Pertussis Pneumococci HiB Influenza Meningococci Hepatitis A Hepatitis B Measlesa Mumpsa Rubellaa Varicellaa
Acute leukemias BIIt BIIt BIIt AIIt CIII AIItu CIII BIIt AIII BIIt BIIt BIIt CIII
Lymphoma, myeloma, MPN AIIt AIIt AIIt AIIt CIIt AIIt CIII BIIt BIIt BIIt BIIt BIIt CIII
Solid tumors AIIt AIIt AIIt AIIt CIII AIIu CIII BIIt BIIt BIIt BIIt BIIt CIII

Strength of recommendation and quality of evidence according to the ESCMID/ECMM grading system is given for each disease—vaccination pair. Further details, especially regarding scheduling of vaccination and therapeutic interventions, are given in the text.

a

Vaccination with live vaccines during immunocompromising therapy is strongly discouraged (DIIt).

HiB, Haemophilus influenzae type B; MPN, myeloproliferative neoplasms.

Acute leukemia

Immunosuppression due to acute leukemia (AL) is most relevant during induction and consolidation therapy. Response to immunization is affected and concerns primary immunization more than booster response. Live vaccines are contraindicated in the immunocompromised patient (DIIt); inactivated, non-conjugated vaccines may not induce sufficient antibody production.

Inactivated vaccines

Protection against diphtheria, tetanus and pertussis (DTP) is impaired in patients with AL [2]. Data on immune response and antibody production to DTP vaccination are only available in children on maintenance and after completion of therapy, showing an antibody response comparable to healthy children [3–5]. We therefore recommend a DTP booster immunization for these patients. They should be vaccinated before starting treatment and after end of therapy if the immune system is reconstituted (BIII).

Influenza increases morbidity and mortality in cancer patients [6–8]. Therefore, all patients >6 months with leukemia should be vaccinated annually against influenza (AIItu) [7, 9, 10]. Patients receiving rituximab should receive the vaccine ≥6 months after therapy because of poor immune response (BIIu) [11]. Data for children with AL show reduced immune response to influenza vaccination [12, 13]. Two doses of influenza vaccine may be more immunogenic than one and are well tolerated in children and young adults [14]. Live-attenuated influenza vaccine cannot be recommended (DIIt).

AL patients should receive antipneumococcal vaccination before treatment (AIIt). The conjugated 13-valent vaccine PCV13 should be administered first; if a patient has not received prior antipneumococcal vaccination, he should be revaccinated with polysaccharide vaccine PPSV23 8–12 weeks later. Priming with a conjugated vaccine may be considered in AL patients as it improves the response to PPSV23 in patients with Hodgkin-lymphoma and patients after bone marrow transplants [15, 16]. If pretreatment vaccination is not feasible, we recommend vaccination after the first chemotherapy cycle and repetition 3 months after chemotherapy, although this strategy has not been specifically evaluated in studies (BIII).

If vaccination against human papilloma virus (HPV) is indicated, vaccination should be performed regardless of immunosuppression; however, immune-response might be reduced [17].

While children with leukemia are at increased risk for Haemophilus influenzae type B (HiB) infections [18], this was not shown for adults except in the context of SCT. Thus, immunization against HiB is only recommended for adults after SCT (CIII) [19].

Patients at risk for hepatitis A virus (HAV) infection, e.g. often reveiving blood products, should be vaccinated [20], but immune-response may be impaired [21]. Patients at high risk of imminent immunosuppression need protection against hepatitis B virus (HBV) [20] and sero-negative patients should be vaccinated [22]. As immune-response for HBV-vaccine was shown to be impaired in this population [23], patients with AL should receive several doses (AIII). Three HBV-vaccinations in AL-patients within 6–12 months after remission was effective in 93.3% of patients vaccinated after bone marrow recovery [24]. Another approach is to combine passive and active prophylaxis with both vaccine and hyperimmunoglobulin [23, 25]. Also, Hepatitis A&B co-immunization is feasible.

Treatment recommendation for rabies is postexposure vaccination [26] with a postexposure double dosage (AIII) due to low immune-response [27]. Pre-exposure vaccination may be indicated for patients with potential occupational exposure (CIII).

There is no general recommendation for vaccination against typhus in patients with AL for lack of data in adults.

Vaccination against tick-born encephalitis may be indicated for patients in endemic areas according to local policy (CIII).

Live vaccines

Vaccination against yellow fever is contra-indicated in AL patients (DIIt) as it may entail severe, life-threatening adverse events like encephalitis [28, 29]. Immunization should not be performed <24 months after immunosuppressive treatment [28].

Measles in immunocompromised patients cause high mortality [30] and, in analogy to children with AL, antibody titers likely decline after chemotherapy [31, 32]. A booster with combined immunization against measles, mumps and rubella (MMR) after completion of treatment is hence recommended for patients with AL (BIIt). This should be performed >24 months after completion of therapy and along serostatus [31].

Adult cancer patients seronegative for varicella zoster virus (VZV) show increased rates of complications (e.g. dissemination, mortality), if primarily infected. Only one study investigated immunity and safety of VZV vaccine in (pediatric) leukemia patients and showed benefits regarding immunity against chickenpox for at least 3 years [33]. Immunization against VZV >24 months after completion of therapy may thus be considered for patients with AL (CIII).

Malignant lymphoma, multiple myeloma and myeloproliferative neoplasms

This section focuses on chemotherapy-treated patients. For vaccination recommendations regarding rituximab-treated patients, refer to ‘Monoclonal antibodies, kinase inhibitors and checkpoint blockade’ section.

Inactivated vaccines

There is solid evidence supporting yearly vaccination of patients with malignant lymphoma, multiple myeloma or myeloproliferative neoplasms with inactivated influenza vaccine to reduce lower respiratory tract infections and hospitalization rates (AIIt) [34–41]. However, there is no conclusive evidence that vaccination also reduces influenza-related mortality [34]. Furthermore, immune-response to single shot vaccination in patients with hematologic malignancies is often impaired [38]. Improved rates of seroconversion were shown after a second shot [35, 37].

A second vaccination against influenza seems reasonable, but can only be recommended with weak evidence (BIIt) and the optimal timing remains unclear. Two small studies suggest improved effectiveness when administering the vaccination directly after a cycle of chemotherapy rather than shortly before the next cycle [42, 43].

Regarding antipneumococcal vaccination, all unvaccinated adult patients should first receive PCV13 followed by PPSV23 vaccine (AIIt) [15, 44, 45]. Vaccination against HiB may be considered in unvaccinated patients (CIIt) [41, 46]. While children are endangered by HiB-induced epiglottitis or meningitis, in adults vaccination aims at preventing HiB-induced lower respiratory tract infection. Vaccination early in the course of disease seems to be associated with superior immune responses [46]. There is little data on vaccination against meningococci apart from asplenia; however, it may be considered in lymphoma patients after chemotherapy (CIII).

Most data available for vaccination against DTP have been obtained from pediatric and allogeneic SCT patients. However, achieving seroprotection is also desirable in adult patients. Therefore vaccination is recommended in case of incomplete vaccination status or requirement of booster vaccination (AIIt) [3, 47, 48].

As insufficient reactivity to tetanus toxoid antigen was reported [48], assessment of antibody titers post vaccination might be considered. Vaccination against HBV is recommended in case of incomplete vaccination status or if a vaccination refreshing is required (BIIt) [23]. Inadequate immune-response should always be considered [23], and in individual patients, passive immunization against HBV could offer short-term protection [23].

Live vaccines

Live vaccines, e.g. against MMR, yellow fever and varicella, are generally contraindicated during chemotherapy (DIIt) [3, 49], including maintenance therapy with monoclonal antibodies or immunomodulatory agents, e.g. lenalidomide, as well as conditions with significant immunosuppression. After completion of immunosuppressive therapy, live vaccines against MMR (BIIt) and varicella (CIII) might be considered after assessment of antibody titers.

Solid tumors

Chemotherapy can induce immune deficiency, including long-term impairment of humoral immunity. Administration of inactivated vaccines in patients with solid tumors is safe while the specific immune response varies and dependends on tumor type and immunosuppression [50].

All adult solid tumor patients should receive yearly vaccination with inactivated influenza vaccine, regardless of age (AIIu). Influenza virus infection increases hospitalization and mortality rates in oncology patients across different tumor types [51]—a population at increased risk of serious influenza-related complications [36]. Additionally, many solid tumor patients are elderly and may benefit from vaccination regardless of cancer. Influenza vaccination has been shown to reduce infection rates and interruption of treatment, possibly contributing to better survival [51–53]. While some studies report a response rate similar to healthy subjects [43, 54–56], the authors of the VACANCE trial suggest a second administration of influenza vaccine in cancer patients as this increased seroconversion from 44% to 73% [57], but further investigations are needed. It was recently shown that cancer patients can be vaccinated independent of chemotherapy and simultaneous administration vaccination and chemotherapy is possible [58].

All unvaccinated adult solid tumour patients should receive antipneumococcal vaccination with PCV-13, followed by PPSV-23 vaccine within 6–12 weeks (AIIt). Vaccinated patients have better survival and are less frequently hospitalized [59]. Response to the vaccine is lower compared with healthy persons [60]. Analogous to findings in Hodgkin’s lymphoma [15], the approach of prime-boost vaccination by PCV13 may provide a better protection for this population, too.

Patients with incomplete vaccination status or indicated refresher vaccination should be vaccinated against DTP. Reportedly, chemotherapy lowers protective antibody titers of diphtheria and pertussis, but not tetanus, in children [47]. However, revaccination is promising: patients showed appropriate antibody response during maintenance treatment and upon completion [3].

Similar to vaccination against DTP, patients with incomplete vaccination status or indicated refresher vaccination should be vaccinated against HBV (BIIt). Older data show immunogenicity and safety of hepatitis B vaccination in patients younger than 60 years under chemotherapy [61]. Assessing titers of diphtheria, tetanus and HBV might be useful after chemotherapy [50].

Vaccination against HAV should be considered (BIIt) as it might prevent fulminant hepatitis or prolonged and relapsing course, potentially delaying therapy. Immunogenicity was demonstrated in children [62], adult HIV patients [63] and post-SCT patients [64]. In addition, STIKO recommends vaccination against HAV for patients receiving transfusions [20], including cancer patients.

Adult patients with malignancies are at increased risk for HiB infections, particularly HiB-induced pneumonia [65]. Immune response to the vaccine is reduced during chemotherapy [65, 66]; therefore, vaccination should be avoided during treatment. Patients vaccinated >14 days before antineoplastic treatment do not require revaccination, unless vaccination status is incomplete or refresher vaccination is indicated. Patients vaccinated during chemotherapy should be revaccinated ≥3 months after therapy. Infection rates have increased in the elderly since establishment of childhood immunization, particularly in those with significant comorbidities with a high fatality rate of invasive infections [67]. Vaccination is recommended if vaccination status is incomplete or vaccination refresher is indicated (CIII).

Vaccination against Meningococcus is routinely recommended for patients with (functional) asplenia. Data for adult solid tumor patients do not exist. There is no rationale to assume increased risk for infections with Meningococcus except for patients with local risk factors such as intrathecal instillation of cytotoxic agents or irradiation of the cranium [68]. Vaccination is only recommended if vaccination status is incomplete or vaccination refresher is indicated (CIII).

In summary, vaccination positively influences hospitalization, morbidity and mortality, but the impact on different solid tumors requires further investigation. However, the aforementioned vaccines should be promoted by treating physicians and discussed with each patient. Further trials are needed to assess the benefit of vaccines in different tumor types taking into account effects of distinct treatments and immune status of patient subgroups.

Intervention-specific vaccination strategies

Certain therapeutic interventions result in profound immunologic challenges and thus require specific vaccination strategies.

Asplenia

Asplenia, both functional and anatomic, is associated with an increased risk of infections by polysaccharide encapsulated bacteria [69]. The risk of severe or fatal infections is also substantially increased, e.g. for fulminant sepsis by pneumococci [70] or HiB [67].

If preoperative vaccination is not possible, vaccination should be performed 14 days after surgery (AI)—longer delay does not yield additional benefit [71], while a shorter wait induces insufficient antibody response. No comparable data are available regarding preoperative vaccination. However, patients should be vaccinated 2 weeks before planned splenectomy at the latest as antibody formation generally takes 9 days [72] (AIII).

In addition to anatomic asplenia, many patients with hematologic diseases should be regarded as functionally asplenic (e.g. all sickle cell patients or patients with hemoglobinopathies) and should thus be vaccinated (AII) [73]. All vaccines recommended in this guideline can be given at once if administered at different injection sites [74].

Patients with anatomic or functional asplenia should first be vaccinated against Streptococcus pneumoniae with PCV13, 6–12 weeks later with PPSV23 [50]. Revaccination should be performed every 6 years (AIIu). While the prime-boost strategy is currently recommended [20], the results of available data vary: benefit was shown for healthy subjects [75] and HIV patients [76], but not for liver/renal transplant recipients [77, 78].

Asplenic patients should be vaccinated with conjugated HiB vaccine (AIIt). A patient is regarded as immunized if he was sequentially vaccinated with the primary series of HiB (by the age of 6 months) and a booster (6–8 months later), or was vaccinated once with HiB vaccine after the age of 14 months. Revaccination is then not needed [79]. While good immunogenicity of conjugated HiB vaccine was repeatedly demonstrated [80–82], further studies showing decreased mortality are needed.

The risk of infections with Neisseria meningitidis is high in asplenic patients and mortality rates range from 40% to 70% [79]. Therefore, these patients should be vaccinated with the tetravalent conjugated meningococcal vaccine, including activity against serogoups A, C, W, Y (MenACWY) (AII), with revaccination every 5 years [83]. Vaccination against serogroup B meningococcus (MenB) is under consideration and currently recommended for use in risk groups (STIKO). MenB was added to the immunization schedule in the United States for those at highest risk—including persons with asplenia—as serogroup B now accounts for 40% of cases [84, 85]. Immunogenicity in asplenic patients has not been shown yet. If the tetravalent conjugate vaccine is not available, a prime-boost strategy (monovalent serogroup C, followed by tetravalent polysaccharide vaccine) is recommended. Scheduled vaccination with MenB vaccine should be re-evaluated and is highly recommended (BII). Measuring of immune response to MenACWY 1 month after administration and—if necessary—a second dose of vaccine should be considered [83, 86].

Patients with asplenia do not have a higher risk of influenza virus infection compared with the general population, but influenza may increase the risk for secondary bacterial pneumonia and sepsis [74, 87]. Thus, the STIKO recommends annual vaccination for all patients with immunodeficiency with influenza vaccine, including asplenic patients [50]. Influenza vaccination was shown to be associated with a 54% reduced mortality compared with unvaccinated asplenic patients [87]. We recommend performing annual vaccination in these patients ≥6 months with inactivated influenza vaccine (AIIt).

Monoclonal antibodies, kinase inhibitors and immune checkpoint blockade

Recent advances in antitumor therapy have significantly improved prognosis of many cancer patients but may pose particular challenges in terms of vulnerability toward infectious agents and success of vaccination strategies. As the field of small-molecule inhibitors and immunotherapy is evolving rapidly, data from large controlled trials regarding safety and efficacy of vaccines in this context are extremely scarce.

Monoclonal antibodies against CD20 result in near complete B cell depletion for up to 6 months after therapy [88]. Although the majority of patients show complete recovery of B cells 1 year after therapy [88], prolonged B cell depletion and hypogammaglobulinemia may occur [89, 90], making vaccination strategies challenging. As a functioning B cell compartment is required for an adequate immuneresponse, vaccination within the first 6 months after anti-CD20 therapy is generally discouraged (DIIu) [56, 91]. Within the first year after anti-CD20 therapy, assessment of antibody titers and revaccination if indicated might be an effective strategy to achieve protective antibody levels despite a diminished immune response (AIIu) [35]. Particularly in case of seasonal influenza vaccine after anti-CD20 therapy, administration of a second booster vaccine has been shown to significantly increase humoral immune response [35]. While most data on vaccination strategies have been obtained with the oldest anti-CD20 antibody rituximab, we believe these recommendations to be transferable to other anti-CD20 antibodies such as ofatumumab or obinutuzumab as well as to other temporarily B cell depleting therapies such as the anti-CD3xCD19 bispecific T-cell engager blinatumomab. As to the vaccination of patients treated with potentially indefinitely B-cell depleting anti-CD19 chimeric antigen receptor T cells, there is virtually no reliable data up to now and decisions regarding vaccination in this setting should be made on an individual basis keeping the patient’s current immune status in mind.

Kinase inhibitors have become standard cancer therapy. Due to the different pathways affected, the effect of kinase inhibitors on immune response to vaccination is entirely dependent on the drug in question. In a small study of patients with metastatic renal cell carcinoma treated with the tyrosine kinase inhibitors sorafenib or sunitinib, no significant difference regarding protective antibody responses after influenza vaccination could be observed compared with healthy controls [92]. Another small study with chronic lymphocytic leukemia patients treated with ibrutinib showed seroprotective titers against common influenza virus strains after vaccination in up to 74% of patients [93]. Inhibitors of the mammalian target of rapamycin such as everolimus or temsirolimus even seem to enhance immune responses [94, 95]. Therefore, decisions on vaccination need to be made on a case-by-case basis. In patients receiving kinase inhibitors, assessment of antibody titers and revaccination (if necessary) is recommended for adequate seroprotection.

Due to their novelty, data on vaccination strategies in patients treated by immune checkpoint blockade, such as anti-PD1, anti-PD-L1 or anti-CTLA4 antibodies, are rare. However, considering their mechanism of action, immune checkpoint inhibitors are likely to enhance rather than diminish immune response and have even been safely explored as vaccine adjuvants [96]. As patients receiving immune checkpoint inhibitors are still at increased risk of infections due to their underlying malignancy, they should receive all appropriate vaccines at the earliest convenience to avoid infectious complications or delay in therapy (BIII).

Autologous stem cell transplantation

Autologous stem cell transplantation (ASCT) following high-dose chemotherapy profoundly impacts the immune system with often persistent reduction of antibody titers to vaccine-preventable diseases requiring a full revaccination to re-establish seroprotection [97, 98]. An overview of recommended vaccines following ASCT is given in Table 3.

Table 3.

Vaccination strategies following autologous stem cell transplantation

Vaccine SoR/QoE Time post-SCT (months) Doses Comment
Influenza AIIt 3–6 1–2 Improved seroprotection with two doses (BIIt)
Pneumococci AIIt 3–6 4 Three doses PCV13 followed by one dose PPSV23
Tetanus BIIu 6–12 3
Diphtheria BIIu 6–12 3 Full dose (“D”) preferred (BIII)
Pertussis CIII 6–12 3 Full dose acellular vaccine (aP) preferred
Poliomyelitis BIIt 6–12 3 Inactivated vaccine only
HiB BIIt 6–12 3–4 Conjugate vaccine preferred
Meningococci BIIt 6–12 1–2 Conjugate vaccine preferred
Hepatitis B BIIt 6–12 3
Measles BIIt 24 1–2 No live vaccine < 24 months post ASCT (DIIt)
Mumps BIIt 24 1–2 No live vaccine < 24 months post ASCT (DIIt)
Rubella BIIt 24 1–2 No live vaccine < 24 months post ASCT (DIIt)
Varicella AIIt 24 1–3 No live vaccine < 24 months post ASCT (DIIt)

Strength of recommendation (SoR) and quality of evidence (QoE) according to the ESCMID grading system is given for each vaccine preventable disease as well as the recommended time point and number of doses of vaccination. Further details are given in the text.

Data on the impact of maintenance therapy after ASCT (e.g. with lenalidomide) on vaccination are scarce. While live-attenuated vaccines must not be administered in this setting (DIIt), inactivated vaccines might be considered.

Starting 3–6 months after ASCT, seasonal vaccination against influenza is recommended (AIIt) [97, 99–101]. ASCT patients are at increased risk of severe courses when developing influenza infection [102, 103]; hence, vaccination early after transplantation might be favored in certain epidemiologic situations. However, immune response to influenza vaccine seems to be significantly impaired within 6 months following ASCT [101], which has to be particularly kept in mind in the setting of prior anti-CD20 therapy [56, 104]. In several small patient series, administration of a second dose of influenza vaccine was shown to significantly increase antibody titers and might therefore be considered in patients following ASCT, especially in case of early vaccination [105, 106].

Despite the lack of a definite head-to-head comparison of the antipneumococcal vaccines PCV13 and PPSV23 in ASCT patients, available data suggest superior immune response to the conjugate vaccine in the setting of an impaired immune system [107]. We therefore recommend, starting 3–6 months after ASCT, administration of three doses of PCV13, each 4–6 weeks apart, followed by one dose of PPSV23 after at least another 8 weeks (AIIt) [97, 108–110].

Vaccination against HiB is recommended 6–12 months after ASCT with administration of at least three doses of conjugated vaccine in order to achieve adequate titers (BIIt) [109, 111–113]. Additionally, vaccination before stem cell harvest may be considered [113]. Against meningococci, the conjugate tetravalent vaccine is preferable and one to two doses are recommended 6–12 months after ASCT according to national endemics (BIIt) [97, 114].

Vaccination with tetanus and diphtheria toxoid is recommended starting 6–12 months after ASCT (BIIu) [97, 98, 109, 114–116]. Administration of three doses of tetanus toxoid usually leads to adequate protective titers [97, 98, 109, 114–116]. In case of diphtheria, vaccination with the full dose of toxoid (D) as licensed for children is preferable to achieve adequate seroprotection (BIII) [97, 98]. Vaccination against poliomyelitis should be performed in three doses only with the inactivated vaccine 6–12 months after ASCT (BIIt) [97, 117, 118]. Regarding pertussis, single vaccination with the lower, adult licensed dosage of acellular pertussis vaccine (ap) seems to be insufficient in adult transplant recipients [119]. Therefore, administration of three full doses acellular pertussis vaccine (aP) are recommended 6–12 months after ASCT (CIII) [97, 120, 121]. Similarly, 6–12 months after ASCT, vaccination with three doses recombinant HBV vaccine is recommended (BIIt) [97, 122]. Patients with a previous history of hepatitis B need to be revaccinated following ASCT (AIIt) [123]. After a full course of HBV vaccination, assessment of antibody titers is recommended to identify nonresponders (CIII) [97].

Current vaccines against MMR, yellow fever and varicella are live-attenuated vaccines and should not be administered during the first 2 years following ASCT or while still undergoing maintenance therapy (DIIt) [3, 49, 97, 124]. After >24 months post-ASCT, administration of live-attenuated vaccines seems to be safe and efficacious, and vaccination against MMR with one to two doses is recommended (BIIt) [97, 114, 115, 125, 126]. In case of measles, assessment of antibody titers before vaccination and administration of vaccine only to seronegative adults might be considered (CIII) [97, 115].

Herpes zoster resulting from reactivation of VZV poses a relevant threat with incidence rates as high as 30% within the first 12 months after ASCT [127]. For seronegative ASCT recipients, primary infection with VZV may pose a significant health hazard. While the risk of herpes zoster seems to be highest within the first year after ASCT [128], early vaccination using a live-attenuated vaccine against VZV has been associated with both low immune response and cases of vaccine virus-induced varicella-like rash [124] and is therefore not recommended, especially given the availability of acyclovir prophylaxis. However, at 2 years after ASCT, vaccination with live-attenuated varicella vaccine seems to be safe [129, 130] and efficient in reducing incidence and severity of herpes zoster in transplant recipients [127, 131]. We recommend vaccination against varicella at >24 months after ASCT using either one dose of Zostavax® [130], licensed for prevention of herpes zoster, or 2 doses of Varilrix® [124, 132], licensed for prevention of primary varicella infection, both containing the live-attenuated Oka strain (BII). In the future, earlier vaccination against varicella might be feasible, as two large phase III trials evaluating a herpes zoster subunit vaccine in older adults showed promising results [133, 134] and a further phase III trial utilizing inactivated varicella vaccine in SCT recipients has already completed enrolment. Inactivated VZV vaccines could be available soon and would possibly change current recommendations.

Vaccinations against other vaccine-preventable diseases depend on local epidemiology and individual risk. Vaccination against HAV seems feasible in ASCT recipients (CIII) and might be coadministered with HBV vaccine [97, 115, 135]. For postexposure prophylaxis, passive immunization is reasonable [97]. In endemic areas, vaccination against tick-borne encephalitis might be recommended according to national policies despite limited data (CIII) [97, 136]. No data on the immunogenicity of HPV in SCT recipients exists, but in adolescents and younger adults, HPV vaccination might be useful (CIII) [97]. Vaccination against travel-associated infectious diseases, such as yellow fever, cholera, typhus, pre-exposure rabies, and Japanese B encephalitis, warrants caution for patients following ASCT, and data on safety and efficacy in these patients are very limited. Decisions regarding vaccination should therefore be only made after thorough risk evaluation for the individual patient. Live-attenuated vaccines (e.g. yellow fever) should not be administered within the first 2 years following ASCT or during maintenance therapy (DIIt) [3, 49, 97, 124].

Following a complete revaccination program, regular assessment of seroprotection against tetanus, diphtheria, poliomyelitis, measles and hepatitis B every 4–5 years is recommendable in patients after ASCT (BIII) [97].

Allogeneic stem cell transplantation

Patients following allogeneic SCT are particularly prone to severe infectious complications. Regarding vaccine-preventable diseases, studies show a decline in protective antibody titers in allogeneic SCT recipients [137]. For these patients, a separate AGIHO guideline exists [138].

Conclusion and outlook

Prevention of infectious diseases by vaccination has proven to be highly effective in many patient populations. This guideline was written to give practical advice for clinicians in hematology and oncology.

As coverage of vaccination is currently suboptimal in the general population as well as in patients at risk, detailed information and clear recommendations by physicians can have a high impact on acceptance of vaccination, especially as the main reasons for denial of vaccination are concerns about interaction with the malignant disease and potential side-effects [139, 140]. Implementation of an institutional vaccination program might further aid in increasing vaccination rates. To eliminate misperception and improve vaccination coverage in the population of cancer patients, educational programs for patients and for physicians focusing on safety and efficacy of vaccines are warranted in the future.

Supplementary Material

Supplementary Table S1

Acknowledgements

The authors would like to thank the “Deutsche Gesellschaft für Hämatologie und Medizinische Onkologie” and the “Arbeitsgemeinschaft Infektionen in der Hämatologie und Onkologie” for their support in establishing this guideline.

Funding

This guideline was in part sponsored by travel grants from the “Deutsche Gesellschaft für Hämatologie und Medizinische Onkologie” (no grant number applies).

Disclosure

DB is consultant to Basilea, Gilead Sciences, Merck Sharp & Dohme/Merck, received research grants from Gilead Sciences and Pfizer, serves on the speakers’ bureau of Astellas, Basilea, Gilead Sciences, Merck Sharp & Dohme/Merck, Pfizer and TEVA, and received travel grants from Astellas, Gliead Sciences, Merck Sharp & Dohme/Merck and Pfizer.

 OAC has received research grants from Actelion, Amplyx, Arsanis, Astellas, AstraZeneca, Basilea, Bayer, Cidara, Duke University (NIH UM1AI104681), F2G, Gilead, GSK, Leeds University, Matinas, Medicines Company, MedPace, Melinta, Merck/MSD, Miltenyi, Pfizer, Rempex, Roche, Sanofi Pasteur, Scynexis, Seres, is a consultant to Amplyx, Actelion, Astellas, Basilea, Cidara, Da Volterra, F2G, Gilead, Janssen, Matinas, Menarini, Merck/MSD, Paratek, PSI, Scynexis, Seres, Summit, Tetraphase, Vical, and received lecture honoraria from Astellas, Basilea, Gilead, Merck/MSD and Pfizer.

 GE received research and travel grants from MSD Sharp and Dohme and Pharmamar; serves on the speakers bureaus of MSD Sharp and Dohme and Gilead Science and is participant of MSD-Advisory Board.

 WJH received research grants from MSD Sharp & Dohme/Merck and Pfizer; serves on the speakers bureaus of Alexion, Astellas, Basilea, Bristol-Myers Squibb, Gilead Sciences, Janssen, MSD Sharp & Dohme and Pfizer; and received travel grants from Alexion, Astellas, Lilly, MSD Sharp & Dohme, Novartis and Pfizer.

 MH served on the speakers’ bureaus for Amgen, Celgene, Janssen, MSD Sharp & Dohme, Novartis and Takeda, and served on advisory boards of Amgen, Celgene, Gilead, Janssen, Sanofi and Takeda.

 MvL-T is supported by the German Federal Ministry of Research and Education (BMBF grants 01EO1002 and 13GW0096D); has received research grants from Pfizer, Novartis, Gilead, Celgene and MSD, is a consultant to Merck/MSD; and received honoraria or travel grants from Basilea, Gilead, Merck/MSD, JanssenCilag and Astellas.

 NL has received speakers’ honoraria from Merck Sharp & Dohme and Roche.

 GM has been a consultant to Gilead and F2G and received honoraria for lectures from Gilead, Pfizer, Basilea and Astellas.

CTR has been a consultant to AbbVie, Astellas Pharma, BMS, Gilead Sciences, Merck Sharp & Dohme, Novartis and Pfizer and served at the speakers’ bureau of Astellas Pharma, Bayer, BMS, Gilead Sciences, Janssen, Merck Sharp & Dohme, Novartis, Pfizer and Roche.

 GS has received honoria from Astallas, Gilead Sciences, GSK, Pfizer, Merck Sharp & Dohme and Neovii.

 MS has received honoraria and served at the speakers’ bureau of Roche.

 MJGTV has served at the speakers’ bureau of Pfizer, Merck, Gilead Sciences and Astellas Pharma, received research funding from 3M, DaVolterra, Astellas Pharma, Gilead Sciences and Merck and is a consultant to Berlin Chemie, Astellas Pharma and Merck. All remaining authors have declared no conflicts of interest.

References

  • 1. Ullmann AJ, Akova M, Herbrecht R. et al. ESCMID* guideline for the diagnosis and management of Candida diseases 2012: adults with haematological malignancies and after haematopoietic stem cell transplantation (HCT). Clin Microbiol Infect 2012; 18: 53–67. [DOI] [PubMed] [Google Scholar]
  • 2. Hamarstrom V, Pauksen K, Svensson H. et al. Tetanus immunity in patients with hematological malignancies. Support Care Cancer 1998; 6(5): 469–472. [DOI] [PubMed] [Google Scholar]
  • 3. Ercan TE, Soycan LY, Apak H. et al. Antibody titers and immune response to diphtheria-tetanus-pertussis and measles-mumps-rubella vaccination in children treated for acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2005; 27(5): 273–277. [DOI] [PubMed] [Google Scholar]
  • 4. Koochakzadeh L, Khosravi MH, Pourakbari B. et al. Assessment of immune response following immunization with DTP/Td and MMR vaccines in children treated for acute lymphoblastic leukemia. Pediatr Hematol Oncol 2014; 31(7): 656–663. [DOI] [PubMed] [Google Scholar]
  • 5. van der Does-van den Berg A, Hermans J, Nagel J, van Steenis G.. Immunity to diphtheria, pertussis, tetanus, and poliomyelitis in children with acute lymphocytic leukemia after cessation of chemotherapy. Pediatrics 1981; 67(2): 222–229. [PubMed] [Google Scholar]
  • 6. Yousuf HM, Englund J, Couch R. et al. Influenza among hospitalized adults with leukemia. Clin Infect Dis 1997; 24(6): 1095–1099. [DOI] [PubMed] [Google Scholar]
  • 7. Engelhard D, Mohty B, de la Camara R. et al. European guidelines for prevention and management of influenza in hematopoietic stem cell transplantation and leukemia patients: summary of ECIL-4 (2011), on behalf of ECIL, a joint venture of EBMT, EORTC, ICHS, and ELN. Transpl Infect Dis 2013; 15(3): 219–232. [DOI] [PubMed] [Google Scholar]
  • 8. Elting LS, Whimbey E, Lo W. et al. Epidemiology of influenza A virus infection in patients with acute or chronic leukemia. Support Care Cancer 1995; 3(3): 198–202. [DOI] [PubMed] [Google Scholar]
  • 9. Ison MG. Epidemiology, prevention, and management of influenza in patients with hematologic malignancy. Infect Disord Drug Targets 2011; 11(1): 34–39. [DOI] [PubMed] [Google Scholar]
  • 10. Casper C, Englund J, Boeckh M.. How I treat influenza in patients with hematologic malignancies. Blood 2010; 115(7): 1331–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Rubin LG, Levin MJ, Ljungman P. et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis 2014; 58(3): e44–100. [DOI] [PubMed] [Google Scholar]
  • 12. Shahgholi E, Ehsani MA, Salamati P. et al. Immunogenicity of trivalent influenza vaccine in children with acute lymphoblastic leukemia during maintenance therapy. Pediatr Blood Cancer 2010; 54(5): 716–720. [DOI] [PubMed] [Google Scholar]
  • 13. Porter CC, Edwards KM, Zhu Y, Frangoul H.. Immune responses to influenza immunization in children receiving maintenance chemotherapy for acute lymphoblastic leukemia. Pediatr Blood Cancer 2004; 42(1): 36–40. [DOI] [PubMed] [Google Scholar]
  • 14. Hakim H, Allison KJ, Van de Velde L-A. et al. Immunogenicity and safety of high-dose trivalent inactivated influenza vaccine compared to standard-dose vaccine in children and young adults with cancer or HIV infection. Vaccine 2016; 34(27): 3141–3148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Chan CY, Molrine DC, George S. et al. Pneumococcal conjugate vaccine primes for antibody responses to polysaccharide pneumococcal vaccine after treatment of Hodgkin's disease. J Infect Dis 1996; 173(1): 256–258. [DOI] [PubMed] [Google Scholar]
  • 16. Kumar D, Chen MH, Welsh B. et al. A randomized, double-blind trial of pneumococcal vaccination in adult allogeneic stem cell transplant donors and recipients. Clin Infect Dis 2007; 45(12): 1576–1582. [DOI] [PubMed] [Google Scholar]
  • 17. Markowitz LE, Dunne EF, Saraiya M. et al. Quadrivalent human papillomavirus vaccine: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2007; 56(Rr-2): 1–24. [PubMed] [Google Scholar]
  • 18. Feldman S, Gigliotti F, Shenep JL. et al. Risk of Haemophilus influenzae type b disease in children with cancer and response of immunocompromised leukemic children to a conjugate vaccine. J Infect Dis 1990; 161(5): 926–931. [DOI] [PubMed] [Google Scholar]
  • 19. Ljungman P. Vaccination of immunocompromised patients. Clin Microbiol Infect 2012; 18: 93–99. [DOI] [PubMed] [Google Scholar]
  • 20. Recommendation of the Ständige Impfkommission (STIKO), Robert Koch-Insitut. 2016; http://www.rki.de/DE/Content/Infekt/EpidBull/Archiv/2016/Ausgaben/34_16.pdf?__blob=publicationFile.
  • 21. Overton ET, Nurutdinova D, Sungkanuparph S. et al. Predictors of immunity after hepatitis A vaccination in HIV-infected persons. J Viral Hepat 2007; 14(3): 189–193. [DOI] [PubMed] [Google Scholar]
  • 22. Sandherr M, Hentrich M, von Lilienfeld-Toal M. et al. Antiviral prophylaxis in patients with solid tumours and haematological malignancies—update of the Guidelines of the Infectious Diseases Working Party (AGIHO) of the German Society for Hematology and Medical Oncology (DGHO). Ann Hematol 2015; 94(9): 1441–1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Goyal S, Pai SK, Kelkar R, Advani SH.. Hepatitis B vaccination in acute lymphoblastic leukemia. Leuk Res 1998; 22(2): 193–195. [DOI] [PubMed] [Google Scholar]
  • 24. Gurina NM, Svedentsov EP, Shardakov VI, Cherepanova VV.. Vaccine prophylaxis of viral hepatitis B in patients with acute leukemia. Ter Arkh 2008; 80(7): 27–29. [PubMed] [Google Scholar]
  • 25. Somjee S, Pai S, Parikh P. et al. Passive active prophylaxis against Hepatitis B in children with acute lymphoblastic leukemia. Leuk Res 2002; 26(11): 989–992. [DOI] [PubMed] [Google Scholar]
  • 26. Wiwanitkit V. Rabies vaccination in a pediatric patient with acute myeloid leukemia during the course of chemotherapy: a case report. Iran J Cancer Prev 2014; 7(2): 105–106. [PMC free article] [PubMed] [Google Scholar]
  • 27. Gibbons RV, Rupprecht CE.. Postexposure rabies prophylaxis in immunosuppressed patients. JAMA 2001; 285(12): 1574–1575. [DOI] [PubMed] [Google Scholar]
  • 28. Bruyand M, Receveur MC, Pistone T. et al. Yellow fever vaccination in non-immunocompetent patients. Med Mal Infect 2008; 38(10): 524–532. [DOI] [PubMed] [Google Scholar]
  • 29. Azevedo LS, Lasmar EP, Contieri FL. et al. Yellow fever vaccination in organ transplanted patients: is it safe? A multicenter study. Transpl Infect Dis 2012; 14(3): 237–241. [DOI] [PubMed] [Google Scholar]
  • 30. Kaplan LJ, Daum RS, Smaron M, McCarthy CA.. Severe measles in immunocompromised patients. JAMA 1992; 267(9): 1237–1241. [PubMed] [Google Scholar]
  • 31. Volc SM, Almeida MT, Abadi MD. et al. Measles and rubella antibody status in children after treatment for acute lymphoblastic leukemia. J Pediatr (Rio J) 2006; 82(6): 481–484. [DOI] [PubMed] [Google Scholar]
  • 32. Feldman S, Andrew M, Norris M. et al. Decline in rates of seropositivity for measles, mumps, and rubella antibodies among previously immunized children treated for acute leukemia. Clin Infect Dis 1998; 27(2): 388–390. [DOI] [PubMed] [Google Scholar]
  • 33. Gershon AA, Steinberg SP.. Persistence of immunity to varicella in children with leukemia immunized with live attenuated varicella vaccine. N Engl J Med 1989; 320(14): 892–897. [DOI] [PubMed] [Google Scholar]
  • 34. Cheuk DK, Chiang AK, Lee TL. et al. Vaccines for prophylaxis of viral infections in patients with hematological malignancies. Cochrane Database Syst Rev 2011; (3): Cd006505. [DOI] [PubMed] [Google Scholar]
  • 35. de Lavallade H, Garland P, Sekine T. et al. Repeated vaccination is required to optimize seroprotection against H1N1 in the immunocompromised host. Haematologica 2011; 96(2): 307–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Eliakim-Raz N, Vinograd I, Zalmanovici Trestioreanu A. et al. Influenza vaccines in immunosuppressed adults with cancer. Cochrane Database Syst Rev 2013; (10): Cd008983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hahn M, Schnitzler P, Schweiger B. et al. Efficacy of single versus boost vaccination against influenza virus in patients with multiple myeloma. Haematologica 2015; 100(7): e285–e288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Mazza JJ, Yale SH, Arrowood JR. et al. Efficacy of the influenza vaccine in patients with malignant lymphoma. Clin Med Res 2005; 3(4): 214–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Musto P, Carotenuto M.. Vaccination against influenza in multiple myeloma. Br J Haematol 1997; 97(2): 505–506. [PubMed] [Google Scholar]
  • 40. Rapezzi D, Sticchi L, Racchi O. et al. Influenza vaccine in chronic lymphoproliferative disorders and multiple myeloma. Eur J Haematol 2003; 70(4): 225–230. [DOI] [PubMed] [Google Scholar]
  • 41. Robertson JD, Nagesh K, Jowitt SN. et al. Immunogenicity of vaccination against influenza, Streptococcus pneumoniae and Haemophilus influenzae type B in patients with multiple myeloma. Br J Cancer 2000; 82(7): 1261–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Meerveld-Eggink A, de Weerdt O, van der Velden AM. et al. Response to influenza virus vaccination during chemotherapy in patients with breast cancer. Ann Oncol 2011; 22(9): 2031–2035. [DOI] [PubMed] [Google Scholar]
  • 43. Wumkes ML, van der Velden AM, Los M. et al. Serum antibody response to influenza virus vaccination during chemotherapy treatment in adult patients with solid tumours. Vaccine 2013; 31(52): 6177–6184. [DOI] [PubMed] [Google Scholar]
  • 44. Bonten MJ, Huijts SM, Bolkenbaas M.. Vaccine against pneumococcal pneumonia in adults. N Engl J Med 2015; 373(1): 91.. [DOI] [PubMed] [Google Scholar]
  • 45. Pasiarski M, Rolinski J, Grywalska E. et al. Antibody and plasmablast response to 13-valent pneumococcal conjugate vaccine in chronic lymphocytic leukemia patients–preliminary report. PLoS One 2014; 9(12): e114966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hartkamp A, Mulder AH, Rijkers GT. et al. Antibody responses to pneumococcal and haemophilus vaccinations in patients with B-cell chronic lymphocytic leukaemia. Vaccine 2001; 19(13-14): 1671–1677. [DOI] [PubMed] [Google Scholar]
  • 47. Reinhardt D, Houliara K, Pekrun A. et al. Impact of conventional chemotherapy on levels of antibodies against vaccine-preventable diseases in children treated for cancer. Scand J Infect Dis 2003; 35(11-12): 851–857. [DOI] [PubMed] [Google Scholar]
  • 48. Sinisalo M, Aittoniemi J, Oivanen P. et al. Response to vaccination against different types of antigens in patients with chronic lymphocytic leukaemia. Br J Haematol 2001; 114(1): 107–110. [DOI] [PubMed] [Google Scholar]
  • 49. Arrowood JR, Hayney MS.. Immunization recommendations for adults with cancer. Ann Pharmacother 2002; 36(7-8): 1219–1229. [DOI] [PubMed] [Google Scholar]
  • 50.Mitteilung der Ständigen Impfkommission (STIKO) am Robert Koch-Institut: Hinweise zu Impfungen für Patienten mit Immundefizienz. 02/2017; 2005.
  • 51. Cooksley CD, Avritscher EB, Bekele BN. et al. Epidemiology and outcomes of serious influenza-related infections in the cancer population. Cancer 2005; 104(3): 618–628. [DOI] [PubMed] [Google Scholar]
  • 52. Earle CC. Influenza vaccination in elderly patients with advanced colorectal cancer. J Clin Oncol 2003; 21(6): 1161–1166. [DOI] [PubMed] [Google Scholar]
  • 53. Wang CS, Wang ST, Lai CT. et al. Impact of influenza vaccination on major cause-specific mortality. Vaccine 2007; 25(7): 1196–1203. [DOI] [PubMed] [Google Scholar]
  • 54. Anderson H, Petrie K, Berrisford C. et al. Seroconversion after influenza vaccination in patients with lung cancer. Br J Cancer 1999; 80(1-2): 219–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Brydak LB, Guzy J, Starzyk J. et al. Humoral immune response after vaccination against influenza in patients with breast cancer. Support Care Cancer 2001; 9(1): 65–68. [DOI] [PubMed] [Google Scholar]
  • 56. Berglund A, Willen L, Grodeberg L. et al. The response to vaccination against influenza A(H1N1) 2009, seasonal influenza and Streptococcus pneumoniae in adult outpatients with ongoing treatment for cancer with and without rituximab. Acta Oncol 2014; 53(9): 1212–1220. [DOI] [PubMed] [Google Scholar]
  • 57. Rousseau B, Loulergue P, Mir O. et al. Immunogenicity and safety of the influenza A H1N1v 2009 vaccine in cancer patients treated with cytotoxic chemotherapy and/or targeted therapy: the VACANCE study. Ann Oncol 2012; 23(2): 450–457. [DOI] [PubMed] [Google Scholar]
  • 58. Keam B, Kim MK, Choi Y. et al. Optimal timing of influenza vaccination during 3-week cytotoxic chemotherapy cycles. Cancer 2017; 123(5): 841–848. [DOI] [PubMed] [Google Scholar]
  • 59. Chiou WY, Hung SK, Lai CL. et al. Effect of 23-valent pneumococcal polysaccharide vaccine inoculated during anti-cancer treatment period in elderly lung cancer patients on community-acquired pneumonia hospitalization: a nationwide population-based cohort study. Medicine (Baltimore) 2015; 94(29): 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nordoy T, Aaberge IS, Husebekk A. et al. Cancer patients undergoing chemotherapy show adequate serological response to vaccinations against influenza virus and Streptococcus pneumoniae. Med Oncol 2002; 19(2): 71–78. [DOI] [PubMed] [Google Scholar]
  • 61. Weitberg AB, Weitzman SA, Watkins E. et al. Immunogenicity of hepatitis B vaccine in oncology patients receiving chemotherapy. J Clin Oncol 1985; 3(5): 718–722. [DOI] [PubMed] [Google Scholar]
  • 62. Köksal Y, Yalçin B, Aydın GB. et al. Immunogenicity of hepatitis a vaccine in children with cancer. Pediatr Hematol Oncol 2006; 23(8): 619–624. [DOI] [PubMed] [Google Scholar]
  • 63. Kourkounti S, Papaizos V, Leuow K. et al. Hepatitis A vaccination and immunological parameters in HIV-infected patients. Viral Immunol 2013; 26(5): 357–363. [DOI] [PubMed] [Google Scholar]
  • 64. Garcia Garrido HM, Wieten RW, Grobusch MP, Goorhuis A.. Response to Hepatitis A vaccination in immunocompromised travelers. J Infect Dis 2015; 212(3): 378–385. [DOI] [PubMed] [Google Scholar]
  • 65. Briere EC, Rubin L, Moro PL. et al. Prevention and control of haemophilus influenzae type b disease: recommendations of the advisory committee on immunization practices (ACIP). MMWR Recomm Rep 2014; 63(Rr-01): 1–14. [PubMed] [Google Scholar]
  • 66. Shenep JL, Feldman S, Gigliotti F. et al. Response of immunocompromised children with solid tumors to a conjugate vaccine for Haemophilus influenzae type b. J Pediatr 1994; 125(4): 581–584. [DOI] [PubMed] [Google Scholar]
  • 67. Singh V, Nanjappa S, Pabbathi S, Greene JN.. Invasive Haemophilus influenzae infection in patients with cancer. Cancer Control 2017; 24(1): 66–71. [DOI] [PubMed] [Google Scholar]
  • 68. Trupl J, Minarik T, Sufliarsky J. et al. Nosocomial bacterial and fungal meningitis in cancer patients. Support Care Cancer 1995; 3(6): 425–427. [DOI] [PubMed] [Google Scholar]
  • 69. Kristinsson SY, Gridley G, Hoover RN. et al. Long-term risks after splenectomy among 8,149 cancer-free American veterans: a cohort study with up to 27 years follow-up. Haematologica 2014; 99(2): 392–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Rubin LG, Schaffner W.. Care of the asplenic patient. N Engl J Med 2014; 371(4): 349–356. [DOI] [PubMed] [Google Scholar]
  • 71. Shatz DV, Romero-Steiner S, Elie CM. et al. Antibody responses in postsplenectomy trauma patients receiving the 23-valent pneumococcal polysaccharide vaccine at 14 versus 28 days postoperatively. J Trauma 2002; 53(6): 1037–1042. [DOI] [PubMed] [Google Scholar]
  • 72. Shatz DV. Vaccination considerations in the asplenic patient. Expert Rev Vaccines 2005; 4(1): 27–34. [DOI] [PubMed] [Google Scholar]
  • 73. Pearson HA, Spencer RP, Cornelius EA.. Functional asplenia in sickle-cell anemia. N Engl J Med 1969; 281(17): 923–926. [DOI] [PubMed] [Google Scholar]
  • 74. Kanhutu K, Jones P, Cheng AC. et al. Spleen Australia guidelines for the prevention of sepsis in patients with asplenia and hyposplenism in Australia and New Zealand. Intern Med J 2017; 47(8): 848–855. [DOI] [PubMed] [Google Scholar]
  • 75. Food and Drug Administration. Vaccines and Related Biological Products Advisory Committee (VRBPAC) adult indication briefing document: Prevnar 13; 2011. www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/bloodvaccinesandotherbiologics/vaccinesandrelatedbiologicalproductsadvisorycomittee/ucm279680.pdf.
  • 76. Lesprit P, Pedrono G, Molina JM. et al. Immunological efficacy of a prime-boost pneumococcal vaccination in HIV-infected adults. AIDS 2007; 21(18): 2425–2434. [DOI] [PubMed] [Google Scholar]
  • 77. Tobudic S, Plunger V, Sunder-Plassmann G. et al. Randomized, single blind, controlled trial to evaluate the prime-boost strategy for pneumococcal vaccination in renal transplant recipients. PLoS One 2012; 7(9): e46133.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kumar D, Chen MH, Wong G. et al. A randomized, double-blind, placebo-controlled trial to evaluate the prime-boost strategy for pneumococcal vaccination in adult liver transplant recipients. Clin Infect Dis 2008; 47(7): 885–892. [DOI] [PubMed] [Google Scholar]
  • 79. Hammerquist RJ, Messerschmidt KA, Pottebaum AA, Hellwig TR.. Vaccinations in asplenic adults. Am J Health Syst Pharm 2016; 73(9): e220–e2e8. [DOI] [PubMed] [Google Scholar]
  • 80. Kristensen K. Antibody response to a Haemophilus influenzae type b polysaccharide tetanus toxoid conjugate vaccine in splenectomized children and adolescents. Scand J Infect Dis 1992; 24(5): 629–632. [DOI] [PubMed] [Google Scholar]
  • 81. Meerveld-Eggink A, de Weerdt O, van Velzen-Blad H. et al. Response to conjugate pneumococcal and Haemophilus influenzae type b vaccines in asplenic patients. Vaccine 2011; 29(4): 675–680. [DOI] [PubMed] [Google Scholar]
  • 82. Mikoluc B, Motkowski R, Kayhty H. et al. Antibody response to Haemophilus influenzae type-b conjugate vaccine in children and young adults with congenital asplenia or after undergoing splenectomy. Eur J Clin Microbiol Infect Dis 2012; 31(5): 805–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Kim DK, Bridges CB, Harriman KH.. Advisory committee on immunization practices recommended immunization schedule for adults aged 19 years or older: United States, 2016. Ann Intern Med 2016; 164(3): 184–194. [DOI] [PubMed] [Google Scholar]
  • 84. Crum-Cianflone N, Sullivan E.. Meningococcal vaccinations. Infect Dis Ther 2016; 5(2): 89–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Cohn AC, MacNeil JR, Harrison LH. et al. Changes in Neisseria meningitidis disease epidemiology in the United States, 1998-2007: implications for prevention of meningococcal disease. Clin Infect Dis 2010; 50(2): 184–191. [DOI] [PubMed] [Google Scholar]
  • 86. Balmer P, Falconer M, McDonald P. et al. Immune response to meningococcal serogroup C conjugate vaccine in asplenic individuals. Infect Immun 2004; 72(1): 332–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Langley JM, Dodds L, Fell D, Langley GR.. Pneumococcal and influenza immunization in asplenic persons: a retrospective population-based cohort study 1990-2002. BMC Infect Dis 2010; 10(1): 219.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. McLaughlin P, Grillo-Lopez AJ, Link BK. et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol 1998; 16(8): 2825–2833. [DOI] [PubMed] [Google Scholar]
  • 89. Nishio M, Fujimoto K, Yamamoto S. et al. Hypogammaglobulinemia with a selective delayed recovery in memory B cells and an impaired isotype expression after rituximab administration as an adjuvant to autologous stem cell transplantation for non-Hodgkin lymphoma. Eur J Haematol 2006; 77(3): 226–232. [DOI] [PubMed] [Google Scholar]
  • 90. Cabanillas F, Liboy I, Pavia O, Rivera E.. High incidence of non-neutropenic infections induced by rituximab plus fludarabine and associated with hypogammaglobulinemia: a frequently unrecognized and easily treatable complication. Ann Oncol 2006; 17(9): 1424–1427. [DOI] [PubMed] [Google Scholar]
  • 91. Bedognetti D, Zoppoli G, Massucco C. et al. Impaired response to influenza vaccine associated with persistent memory B cell depletion in non-Hodgkin's lymphoma patients treated with rituximab-containing regimens. J Immunol (Baltimore, Md: 1950). 2011; 186(10): 6044–6055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Mulder SF, Jacobs JF, Olde Nordkamp MA. et al. Cancer patients treated with sunitinib or sorafenib have sufficient antibody and cellular immune responses to warrant influenza vaccination. Clin Cancer Res 2011; 17(13): 4541–4549. [DOI] [PubMed] [Google Scholar]
  • 93. Sun C, Gao J, Couzens L. et al. Seasonal influenza vaccination in patients with chronic lymphocytic leukemia treated with ibrutinib. JAMA Oncol 2016; 2(12): 1656–1657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Mannick JB, Del Giudice G, Lattanzi M. et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med 2014; 6(268): 268ra179.. [DOI] [PubMed] [Google Scholar]
  • 95. Struijk GH, Minnee RC, Koch SD. et al. Maintenance immunosuppressive therapy with everolimus preserves humoral immune responses. Kidney Int 2010; 78(9): 934–940. [DOI] [PubMed] [Google Scholar]
  • 96. Madan RA, Mohebtash M, Arlen PM. et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13(5): 501–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Ljungman P, Cordonnier C, Einsele H. et al. Vaccination of hematopoietic cell transplant recipients. Bone Marrow Transplant 2009; 44(8): 521–526. [DOI] [PubMed] [Google Scholar]
  • 98. Nordoy T, Husebekk A, Aaberge IS. et al. Humoral immunity to viral and bacterial antigens in lymphoma patients 4-10 years after high-dose therapy with ABMT. Serological responses to revaccinations according to EBMT guidelines. Bone Marrow Transplant 2001; 28(7): 681–687. [DOI] [PubMed] [Google Scholar]
  • 99. Gandhi MK, Egner W, Sizer L. et al. Antibody responses to vaccinations given within the first two years after transplant are similar between autologous peripheral blood stem cell and bone marrow transplant recipients. Bone Marrow Transplant 2001; 28(8): 775–781. [DOI] [PubMed] [Google Scholar]
  • 100. Pauksen K, Linde A, Hammarstrom V. et al. Granulocyte-macrophage colony-stimulating factor as immunomodulating factor together with influenza vaccination in stem cell transplant patients. Clin Infect Dis 2000; 30(2): 342–348. [DOI] [PubMed] [Google Scholar]
  • 101. Engelhard D, Nagler A, Hardan I. et al. Antibody response to a two-dose regimen of influenza vaccine in allogeneic T cell-depleted and autologous BMT recipients. Bone Marrow Transplant 1993; 11(1): 1–5. [PubMed] [Google Scholar]
  • 102. Ljungman P, de la Camara R, Perez-Bercoff L. et al. Outcome of pandemic H1N1 infections in hematopoietic stem cell transplant recipients. Haematologica 2011; 96(8): 1231–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Nichols WG, Guthrie KA, Corey L, Boeckh M.. Influenza infections after hematopoietic stem cell transplantation: risk factors, mortality, and the effect of antiviral therapy. Clin Infect Dis 2004; 39(9): 1300–1306. [DOI] [PubMed] [Google Scholar]
  • 104. Villa D, Gubbay J, Sutherland DR. et al. Evaluation of 2009 pandemic H1N1 influenza vaccination in adults with lymphoid malignancies receiving chemotherapy or following autologous stem cell transplant. Leuk Lymphoma 2013; 54(7): 1387–1395. [DOI] [PubMed] [Google Scholar]
  • 105. Gueller S, Allwinn R, Mousset S. et al. Enhanced immune response after a second dose of an AS03-adjuvanted H1N1 influenza A vaccine in patients after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2011; 17(10): 1546–1550. [DOI] [PubMed] [Google Scholar]
  • 106. Engelhard D, Zakay-Rones Z, Shapira MY. et al. The humoral immune response of hematopoietic stem cell transplantation recipients to AS03-adjuvanted A/California/7/2009 (H1N1)v-like virus vaccine during the 2009 pandemic. Vaccine 2011; 29(9): 1777–1782. [DOI] [PubMed] [Google Scholar]
  • 107. Juergens C, de Villiers PJ, Moodley K. et al. Safety and immunogenicity of 13-valent pneumococcal conjugate vaccine formulations with and without aluminum phosphate and comparison of the formulation of choice with 23-valent pneumococcal polysaccharide vaccine in elderly adults: a randomized open-label trial. Hum Vaccin Immunother 2014; 10(5): 1343–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Cordonnier C, Labopin M, Chesnel V. et al. Immune response to the 23-valent polysaccharide pneumococcal vaccine after the 7-valent conjugate vaccine in allogeneic stem cell transplant recipients: results from the EBMT IDWP01 trial. Vaccine 2010; 28(15): 2730–2734. [DOI] [PubMed] [Google Scholar]
  • 109. van der Velden AM, Claessen AM, van Velzen-Blad H. et al. Vaccination responses and lymphocyte subsets after autologous stem cell transplantation. Vaccine 2007; 25(51): 8512–8517. [DOI] [PubMed] [Google Scholar]
  • 110. Antin JH, Guinan EC, Avigan D. et al. Protective antibody responses to pneumococcal conjugate vaccine after autologous hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2005; 11(3): 213–222. [DOI] [PubMed] [Google Scholar]
  • 111. van der Velden AM, Claessen AM, van Velzen-Blad H. et al. Development of functional Haemophilus influenzae type b antibodies after vaccination. Haematologica 2005; 90(11): 1582–1584. [PubMed] [Google Scholar]
  • 112. Chan CY, Molrine DC, Antin JH. et al. Antibody responses to tetanus toxoid and Haemophilus influenzae type b conjugate vaccines following autologous peripheral blood stem cell transplantation (PBSCT). Bone Marrow Transplant 1997; 20(1): 33–38. [DOI] [PubMed] [Google Scholar]
  • 113. Molrine DC, Guinan EC, Antin JH. et al. Haemophilus influenzae type b (HIB)-conjugate immunization before bone marrow harvest in autologous bone marrow transplantation. Bone Marrow Transplant. 1996; 17(6): 1149–1155. [PubMed] [Google Scholar]
  • 114. Patel SR, Ortin M, Cohen BJ. et al. Revaccination with measles, tetanus, poliovirus, Haemophilus influenzae type B, meningococcus C, and pneumococcus vaccines in children after hematopoietic stem cell transplantation. Clin Infect Dis 2007; 44(5): 625–634. [DOI] [PubMed] [Google Scholar]
  • 115. Tomblyn M, Chiller T, Einsele H. et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: a global perspective. Biol Blood Marrow Transplant 2009; 15(10): 1143–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Hammarstrom V, Pauksen K, Bjorkstrand B. et al. Tetanus immunity in autologous bone marrow and blood stem cell transplant recipients. Bone Marrow Transplant 1998; 22(1): 67–71. [DOI] [PubMed] [Google Scholar]
  • 117. Ljungman P, Aschan J, Gustafsson B. et al. Long-term immunity to poliovirus after vaccination of allogeneic stem cell transplant recipients. Bone Marrow Transplant 2004; 34(12): 1067–1069. [DOI] [PubMed] [Google Scholar]
  • 118. Engelhard D, Handsher R, Naparstek E. et al. Immune response to polio vaccination in bone marrow transplant recipients. Bone Marrow Transplant 1991; 8(4): 295–300. [PubMed] [Google Scholar]
  • 119. Small TN, Zelenetz AD, Noy A. et al. Pertussis immunity and response to tetanus-reduced diphtheria-reduced pertussis vaccine (Tdap) after autologous peripheral blood stem cell transplantation. Biol Blood Marrow Transplant 2009; 15(12): 1538–1542. [DOI] [PubMed] [Google Scholar]
  • 120. Edwards KM, Berbers GA.. Immune responses to pertussis vaccines and disease. J Infect Dis 2014; 209 Suppl 1: S10–S15. [DOI] [PubMed] [Google Scholar]
  • 121. Inaba H, Hartford CM, Pei D. et al. Longitudinal analysis of antibody response to immunization in paediatric survivors after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2012; 156(1): 109–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Jaffe D, Papadopoulos EB, Young JW. et al. Immunogenicity of recombinant hepatitis B vaccine (rHBV) in recipients of unrelated or related allogeneic hematopoietic cell (HC) transplants. Blood 2006; 108(7): 2470–2475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Onozawa M, Hashino S, Darmanin S. et al. HB Vaccination in the prevention of viral reactivation in allogeneic hematopoietic stem cell transplantation recipients with previous HBV infection. Biol Blood Marrow Transplant 2008; 14(11): 1226–1230. [DOI] [PubMed] [Google Scholar]
  • 124. Sasadeusz J, Prince HM, Schwarer A. et al. Immunogenicity and safety of a two-dose live attenuated varicella vaccine given to adults following autologous hematopoietic stem cell transplantation. Transpl Infect Dis 2014; 16(6): 1024–1031. [DOI] [PubMed] [Google Scholar]
  • 125. Spoulou V, Giannaki M, Vounatsou M. et al. Long-term immunity to measles, mumps and rubella after MMR vaccination among children with bone marrow transplants. Bone Marrow Transplant 2004; 33(12): 1187–1190. [DOI] [PubMed] [Google Scholar]
  • 126. Ljungman P, Fridell E, Lonnqvist B. et al. Efficacy and safety of vaccination of marrow transplant recipients with a live attenuated measles, mumps, and rubella vaccine. J Infect Dis 1989; 159(4): 610–615. [DOI] [PubMed] [Google Scholar]
  • 127. Hata A, Asanuma H, Rinki M. et al. Use of an inactivated varicella vaccine in recipients of hematopoietic-cell transplants. N Engl J Med 2002; 347(1): 26–34. [DOI] [PubMed] [Google Scholar]
  • 128. Rogers JE, Cumpston A, Newton M, Craig M.. Onset and complications of varicella zoster reactivation in the autologous hematopoietic cell transplant population. Transpl Infect Dis 2011; 13(5): 480–484. [DOI] [PubMed] [Google Scholar]
  • 129. Aoki T, Koh K, Kawano Y. et al. Safety of live attenuated high-titer varicella-zoster virus vaccine in pediatric allogeneic hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant 2016; 22(4): 771–775. [DOI] [PubMed] [Google Scholar]
  • 130. Issa NC, Marty FM, Leblebjian H. et al. Live attenuated varicella-zoster vaccine in hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant 2014; 20(2): 285–287. [DOI] [PubMed] [Google Scholar]
  • 131. Redman RL, Nader S, Zerboni L. et al. Early reconstitution of immunity and decreased severity of herpes zoster in bone marrow transplant recipients immunized with inactivated varicella vaccine. J Infect Dis 1997; 176(3): 578–585. [DOI] [PubMed] [Google Scholar]
  • 132. Leung TF, Li CK, Hung EC. et al. Immunogenicity of a two-dose regime of varicella vaccine in children with cancers. Eur J Haematol 2004; 72(5): 353–357. [DOI] [PubMed] [Google Scholar]
  • 133. Cunningham AL, Lal H, Kovac M. et al. Efficacy of the herpes zoster subunit vaccine in adults 70 years of age or older. N Engl J Med 2016; 375(11): 1019–1032. [DOI] [PubMed] [Google Scholar]
  • 134. Lal H, Cunningham AL, Godeaux O. et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med 2015; 372(22): 2087–2096. [DOI] [PubMed] [Google Scholar]
  • 135. Tung J, Carlisle E, Smieja M. et al. A randomized clinical trial of immunization with combined hepatitis A and B versus hepatitis B alone for hepatitis B seroprotection in hemodialysis patients. Am J Kidney Dis 2010; 56(4): 713–719. [DOI] [PubMed] [Google Scholar]
  • 136. Dengler TJ, Zimmermann R, Meyer J. et al. Vaccination against tick-borne encephalitis under therapeutic immunosuppression. Reduced efficacy in heart transplant recipients. Vaccine 1999; 17(7-8): 867–874. [DOI] [PubMed] [Google Scholar]
  • 137. Seggewiss R, Einsele H.. Immune reconstitution after allogeneic transplantation and expanding options for immunomodulation: an update. Blood 2010; 115(19): 3861–3868. [DOI] [PubMed] [Google Scholar]
  • 138. Ullmann AJ, Schmidt-Hieber M, Bertz H. et al. Infectious diseases in allogeneic haematopoietic stem cell transplantation: prevention and prophylaxis strategy guidelines 2016. Ann Hematol 2016; 95: 1435–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Poeppl W, Lagler H, Raderer M. et al. Influenza vaccination perception and coverage among patients with malignant disease. Vaccine 2015; 33(14): 1682–1687. [DOI] [PubMed] [Google Scholar]
  • 140. Urun Y, Akbulut H, Demirkazik A. et al. Perception about influenza and pneumococcal vaccines and vaccination coverage among patients with malignancies and their family members. J Buon 2013; 18(2): 511–515. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Table S1

Articles from Annals of Oncology are provided here courtesy of Oxford University Press

RESOURCES