Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Sep 24, 2023; 14(9): 343-356
Published online Sep 24, 2023. doi: 10.5306/wjco.v14.i9.343
Effectiveness and safety of COVID-19 vaccines in patients with oncological diseases: State-of-the-art
Nedelcho Ivanov, Department of Clinical Immunology with Stem Cell Bank, University Hospital Alexanrovska, Sofia 1431, Bulgaria
Boris Krastev, Medical Center Nadezhda, Medical Center Nadezhda, Sofia 1407, Bulgaria
Dimitrina Georgieva Miteva, Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
Hristiana Batselova, Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital St. George, Plovdiv 6000, Bulgaria
Radostina Alexandrova, Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia 1000, Bulgaria
Tsvetelina Velikova, Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
ORCID number: Nedelcho Ivanov (0000-0002-7471-7368); Boris Krastev (0000-0003-4196-0828); Dimitrina Georgieva Miteva (0000-0002-5931-2426); Hristiana Batselova (0000-0002-6201-848X); Radostina Alexandrova (0000-0002-7699-6479); Tsvetelina Velikova (0000-0002-0593-1272).
Author contributions: Ivanov N and Velikova T contributed to conceptualization; Krastev B contributed to methodology; Miteva DG contributed to software; Ivanov N, Alexandrova R, and Velikova T contributed to validation; Ivanov N contributed to formal analysis; Krastev B contributed to investigation; Batselova H contributed to resources; Miteva DG contributed to data curation; Ivanov N, Krastev B, and Miteva DG contributed to writing-original draft preparation; Batselova H contributed to writing-review & editing; Miteva DG contributed to visualization; Velikova T contributed to supervision, project administration, and funding acquisition; and all authors revised and approved the final version of the manuscript.
Supported by the European Union-Next Generation EU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, No. BG-RRP-2.004-0008.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nedelcho Ivanov, FRCS (Hon), MD, PhD, Assistant Professor, Department of Clinical Immunology with Stem Cell Bank, University Hospital Alexanrovska, St. Georfi Sofiiski 1 str, Sofia 1431, Bulgaria. nedelcho.ivanov93@gmail.com
Received: June 28, 2023
Peer-review started: June 28, 2023
First decision: July 6, 2023
Revised: August 6, 2023
Accepted: September 12, 2023
Article in press: September 12, 2023
Published online: September 24, 2023
Processing time: 83 Days and 5.4 Hours

Abstract

Although the coronavirus disease 2019 (COVID-19) pandemic was declared to be no longer “a public health emergency of international concern” with its wide range of clinical manifestations and late complications, severe acute respiratory syndrome coronavirus 2 infection proved to be a serious threat, especially to the elderly and patients with comorbidities. Patients with oncologic diseases are vulnerable to severe infection and death. Indeed, patients with oncohematological diseases have a higher risk of severe COVID-19 and impaired post-vaccination immunity. Unfortunately, cancer patients are usually excluded from vaccine trials and investigations of post-vaccinal immune responses and the effectiveness of the vaccines. We aimed to elucidate to what extent patients with cancer are at increased risk of developing severe COVID-19 and what is their overall case fatality rate. We also present the current concept and evidence on the effectiveness and safety of COVID-19 vaccines, including boosters, in oncology patients. In conclusion, despite the considerably higher mortality in the cancer patient group than the general population, countries with high vaccination rates have demonstrated trends toward improved survival of cancer patients early and late in the pandemic.

Key Words: COVID-19, COVID-19 vaccines, RNA vaccines, Cancer, Oncological, Safety, Efficacy, Immunogenicity

Core Tip: The coronavirus disease 2019 (COVID-19) pandemic has greatly impacted the lives of cancer patients. Their medical care has been challenging, given the competing risks of death from cancer and serious complications from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Cancer patients are at high risk of severe complications and death from COVID-19. Protective SARS-CoV-2 antibodies and cellular immune response are induced after infection or/and COVID-19 vaccination. Vaccines decrease the risk of hospitalization and death from COVID-19. Therefore, vaccination of specific vulnerable groups, such as oncological patients, and all people in general, will slow the virus spread and save lives.



INTRODUCTION

The coronavirus disease 2019 (COVID-19) pandemic has considerably impacted the lives of cancer patients. Their medical care has been challenging because of the competing risks of death from cancer or serious complications from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the likely higher lethality in immunocompromised hosts[1,2]. Furthermore, patients diagnosed with malignancies are at higher risk of developing severe COVID-19[3] and fatal outcomes due to the disease. Studies have demonstrated variable mortality rates among subjects with hematological cancers and solid tumors, with some reporting fatality cases of as much as 40% of the infected subjects[4]. Despite this considerably higher mortality than the one observed in the general population, trends towards improved survival during the evolution of the pandemic have already been demonstrated in Europe, and much of this could be a direct result of the rigorous COVID-19 vaccination in this region[5].

Since the beginning of the pandemic, hundreds of different therapeutic options have been studied, including those well-known in the treatment of other diseases, such as reoriented drugs. Amongst them are remdesivir (initially developed for hepatitis C treatment, tocilizumab-rheumatoid arthritis, hydroxychloroquine-malaria, lupus, etc), corticosteroids, plasma from donors who have recovered from COVID-19, monoclonal antibodies (casirivimab + imdevimab, bamlanivimab, sotrovimab, cilgavimab + tixagevimab, etc), Janus kinase inhibitors (baricitinib), and even mesenchymal stem cells[6,7]. Targeting both the virus itself and the host’s immune response with variable effectiveness during the different stages of the disease. However, prevention in the form of COVID vaccines remains the most desirable option for the general population both in long-term health-related and financial terms. Cancer patients are no exception in this regard. But exactly how effective are vaccines in cancer patients compared to the general population? This is the question we will try to answer.

In this review, we elucidated to what extent patients with cancer are at increased risk of developing severe COVID-19 and what is their overall case fatality rate. We also present the current concept and evidence on the effectiveness and safety of COVID-19 vaccines, including boosters, in oncology patients.

SEARCH STRATEGY

We performed a modified form of a biomedical narrative review according to recent recommendations for writing[8]. First, we thoroughly searched the scientific bibliographic databases Medline (PubMed) and Scopus. We used relevant free-text and Medical Subject Headings terms, as follows: (“COVID-19” OR “SARS-CoV-2”) AND (“cancer patients” OR “oncological patients”) AND (“COVID-19 vaccine” OR “mRNA vaccine”). We confined the search from January 1, 2020 to June 20, 2023. Then we identified additional papers using the search engine Google Scholar. Information from advisory committee meetings was also added.

COVID-19 AND PATIENTS WITH ONCOLOGICAL DISEASES

Patients with oncologic diseases are affected by SARS-CoV-2 in many different ways. Similar to many other infections, COVID-19 poses an additional risk of a fatal outcome for cancer patients. However, it is challenging to say to what extent patients with malignancies are threatened by complications of severe infections. As oncological diseases and treatment protocols are extremely diverse, it can be expected that the course of SARS-CoV2 infection would also be quite different[9-11].

The stage of disease, type of malignancy, and the sort and phase of the applied treatment modalities (surgery, chemotherapy, radiation therapy, and immunotherapy) introduce even more variables and more superimposing confounding factors, making this group of patients even more heterogeneous and difficult for overall risk assessment. Cancer patients who have recently undergone surgery or chemotherapy (especially during the induction phase with high-dose intensive regimens) are at a dramatically increased risk of death from COVID[12,13]. Side effects of chemotherapy, such as secondary immunodeficiency due to severe leukopenia and specific tissue toxicity due to some chemo- and immunotherapeutics, can significantly alter the course of COVID-19 infection, from worsening the patient’s overall condition and increasing the risk of complications and death to masking or mimicking the radiological pulmonary signs (e.g., immune checkpoint related pneumonitis)[13]. Finally, another confounding factor is the various therapeutic regimens used to treat infection in hospitals and intensive care units worldwide. Cancer patients are treated as high risk by default, which carries a risk (polypharmacy, drug interactions, adverse drug effects, acute kidney or liver failure, etc)[14].

Below, we present data from several studies that attempt to measure and objectify this risk. The first large-scale meta-analysis by 2020 done by Zhang et al[11] of 15 studies involving a total of 3019 patients from Europe, the United Kingdom, the United States, Canada, and Asia detected 22.4% circulating free RNAs (CFRs) in cancer patients with COVID-19, compared to 5.9% in noncancer patients. As in other patients, risk factors influencing the course and mortality are: Being over 65 years old, male sex, and having comorbidities (especially hypertension and diabetes). No significant difference in mortality was found between different continents. The study found that mortality in patients with lung cancer and hematological malignancies was highest, although the incidence of complications did not differ[11].

A study by Yang et al[15] involving 1575 patients, of whom 52 with various cancers (lung, colorectal, breast, cervical, thyroid, etc) showed that oncologic patients are at higher risk of presenting as severe/critical cases and are more likely to develop acute respiratory distress syndrome. Also, other life-threatening complications such as myocardial infarction and shock are significantly increased in frequency. Lower lymphocyte count, as well as higher concentrations of C-reactive protein, D-dimer, procalcitonin, interleukin 6 (IL-6), and lactate dehydrogenase, were reported to reach P < 0.05. Cancer patients are also more likely to have comorbidities, which, as it becomes clear in this study, contributes seriously to the overall higher CFR[15].

A meta-analysis of 122 papers and 9 studies, including a total of 805 patients by Afshar et al[16], demonstrated how heterogeneous the data on mortality in cancer patients are. They showed that cancer patients are more likely to be admitted to intensive care units, need invasive ventilation, and are more likely to die. The published CFR in the analyzed studies ranges from 5.5% to 60.0%, with a pooled CFR of 21%. However, the authors warn that these data should be interpreted cautiously due to the high heterogeneity and the small number of patients in most studies[16].

Large-scale survival analysis by Li et al[9] based on data from United Kingdom Biobank followed 4606 cancer patients (288 positives) and 4606 noncancer patients (275 positives) for 21 mo after the SARS-CoV2 test. The cumulative CFR of the positive cancer patients was six times higher than the negative ones. The hazard ratio was assessed for each specific malignancy in the study, and the results showed that hematological malignancies, melanoma, kidney, and uterine cancer had particularly high CFRs (up to 10 times higher than the noncancer controls). The authors emphasize the importance of timely vaccination in these groups of patients[9].

In contrast to the data above, a study by Brar et al[17] included 585 patients, 117 with active malignancies. It showed no statistically significant difference in morbidity or mortality in cancer patients vs the general population. Furthermore, the authors argued that the studies claiming the opposite did not consider confounding factors such as age, sex, and comorbidities. According to this study, cytotoxic treatment within 90 d of admission is not associated with worse outcomes[17].

A team from London published a study in onco-hematology patients, where 40% (14 of 35) of patients hospitalized with COVID-19 had succumbed to the infection[18]. In general, COVID-19 appears to have an increased risk of complications and mortality in a large proportion of cancer patients. In addition, besides the virus itself, the pandemic and the restrictive measures were associated with disrupted access to medical care, hindered timely diagnosis and treatment, the lack of follow-up of many patients and lower quality of life[19]. Studies have shown that since the beginning of the pandemic, the total number of newly diagnosed cancers has dropped substantially[20]. As many authors warned, this inevitably led to an increased frequency of advanced cancers at diagnosis. Delaying diagnosis and treatment resulted in lower chances of survival[21]. Yong et al[22] conducted a study in Canada using microsimulation models, which estimated that for colorectal cancers, suspending primary screening for only 6 mo will increase cancer incidence by about 2200 cases, of which about 960 will be lethal over time. Consequences that otherwise would be prevented by the screening program and early detection.

Furthermore, there are many other indirect ways the COVID-19 pandemic affects cancer patients’ quality of life and mortality[13]. At the same time, the standard of living, the structure and stability of the health care system, and even political factors in connection with dealing with the pandemic play roles that should not be underestimated[23]. Knowing risk factors for the severity and mortality of COVID-19, cancer patients have their unique risk factors. They may include active and progressing cancer, type of cancer, administration of cytotoxic chemotherapy, radiation therapy, impaired immune system due to leukocytopenia, low immunoglobulin levels, long-lasting immunosuppression, comorbidities, and others.

Malignancies reported as comorbidities in patients hospitalized with confirmed COVID-19 in different countries are: (1) Malignancies in 7.2% in a cohort study with 138 adults with confirmed COVID-19 pneumonia in Wuhan, China, in January 2020[24]; (2) malignancies in 8% at admission in a cohort study with 1591 patients with laboratory-confirmed COVID-19 in Lombardy, Italy between February 20 and March 18, 2020[25]; and (3) Malignancies reported in 5.6% at admission in a cohort study with 5700 patients with confirmed COVID-19 infection hospitalized in 12 New York City hospitals between March 1 and April 4, 2020[26].

In a cohort study of 928 adults with COVID-19 and current or past cancer diagnosis, solid tumors were found in 82%, including breast (21%), hematologic (22%), prostate (16%), gastrointestinal (12%), thoracic (10%), gynecologic (5%), and renal cell carcinoma (5%)[27,28]. The estimated overall mortality in the research was 13%: 20% for patients with multiple cancers, 18% for patients with hematological malignancies, and 12% for patients with solid tumors[27].

Zhang et al[11] showed the COVID-19 fatality rates in subgroup analysis: (1) By cancer type: 32.9% in patients with lung cancer; 34.2% in patients with hematologic cancer; 17.2% in patients with solid cancer; and (2) By cancer treatment: 25.6% in patients with chemotherapy, 27.6% in patients with surgery, 24.3% in patients with immunotherapy, 21.3% in patients with targeted therapy, and 20.5% in patients with radiation therapy[11].

Children with cancer and positive for COVID-19 are at higher risk of severe illness than children without cancer. The cohort study found that about 20% of pediatric cancer patients with COVID-19 experienced a severe infection, compared to 1%-6% of children in the general population[29]. Among patients with hematologic malignancy and laboratory-confirmed COVID-19, mortality was reported in 34% of adults and 4% of children[4].

We can summarize that the main challenges in cancer patients regarding COVID-19 are the often immunocompromised state (e.g., due to leukocytopenia, low immunoglobulin levels, long-lasting immunosuppression), the treatment (e.g., severe chemotherapy, radiation therapy), progression of cancer, comorbidities, and others.

IMMUNE RESPONSE IN CANCER PATIENTS

Cancer cells induce an immune suppressive microenvironment and use various mechanisms to “escape” the body’s immune response. As a systemic disease, cancer causes a wide range of functional and compositional changes in the immune system and can affect the body’s defenses against various pathogens[30,31].

Dendritic cells (DCs) are antigen-presenting cells with an essential role in originating and directing cellular and humoral immune responses, converging innate and adaptive immunity. DCs have been recognized as the most potent professional antigen-presenting cells[32].

Tumors use different strategies to alter DC maturation and function, such as: (1) The ability to influence the capacity of hematopoietic progenitor cells to differentiate into functional DCs[33,34]; (2) production of various immunosuppressive factors that block the maturation of CD34+ stem cells into DCs[35]; and (3) spontaneous apoptosis of DCs in peripheral blood of patients with breast cancer[36]. Quantitative and functional DC deficiencies have been widely observed in patients with several types of cancer including breast cancer[37,38], prostate cancer[38], non-small cell lung cancer[39,40], colon cancer[41], and melanoma[42].

Data have revealed that tumors disrupt normal hematopoiesis, leading to extramedullary hematopoiesis and myeloid skewing. The three branches of terminally differentiated myeloid cells (macrophages, DCs, and granulocytes) are essential for normal innate and adaptive immune response functioning. The tumor microenvironment alters myeloid cells and can convert them into potent immunosuppressive cells[43,44]. Lymphopenia caused by disease or treatment is frequent in oncology patients and affects their prognosis[45,46].

T cells, one of the primary arms of the adaptive immune response, are also affected in oncology patients. Cancer cells express various membrane and soluble T-cell inhibitory signals. For example, programmed cell death protein-ligand 1 linking to programmed cell death protein 1 on T cells results in decreased activation, proliferation, survival, and cytotoxicity[47]. The last discovery led to the development of checkpoint inhibitors, a breakthrough in immuno-oncology, which led to the 2018 Nobel Prize for Physiology or Medicine. Indoleamine 2,3-dioxygenase, a soluble enzyme physiologically expressed in many tissues, is overproduced in some cancers leading to tryptophan depletion in the tumor microenvironment. T cells, being highly sensitive to tryptophan deprivation, suffer significant functional impairment, promoting tumor growth[48]. An increased rate of CD4+CD25+ regulatory T cells with potent immunosuppressive properties in the peripheral blood of individuals with cancer diseases has been reported[49,50].

Additionally, regulatory B cells (Bregs) are a newly designated subset of B cells that play a central role in regulating immune responses associated with inflammation, autoimmunity, and cancer. Increased Bregs express immunosuppressive properties in gastric cancer through the secretion of anti-inflammatory molecules, such as IL-10, and facilitating the conversion of T cells to regulatory T cells[44,51,52]. Additionally, tumor progression is associated with the dysfunction of natural killer cells due to the combined action of tissue-specific and systemic factors[53]. All of these immune alterations in cancer patients contribute to the differences in immune response after vaccination, including after COVID-19 vaccine administration. Before the COVID-19 pandemic, we had an experience with influenza vaccine administration in patients with oncological diseases. Infectious complications resulting from bacterial, fungal, and viral (often due to reactivation of latent disease, primarily in patients with hematological malignancies) diseases are a severe cause of morbidity and mortality in cancer patients[54]. Oncology patients receiving chemotherapy are at increased risk of influenza virus infection and serious post-influenza complications. Cancer patients are eligible for influenza vaccination, although their response may be suboptimal due to immunosuppression associated with cancer itself and/or its treatment[55,56]. Data have shown that cancer patients receiving chemotherapy can respond to influenza vaccination[57].

Breast cancer patients receiving influenza vaccination during FEC (5-fluorouracil, epirubicin, and cyclophosphamide)-containing treatment regimens have exhibited significantly lower responses to influenza virus vaccination than healthy controls. Vaccination early during the chemotherapy cycle (day 4) induces better responses than vaccination on day 16[58]. The summary of the available evidence reveals that immunization of individuals with malignancies is critical to their care and may protect them from significant morbidity and mortality associated with vaccine-preventable diseases[59].

COVID-19 VACCINES FOR PATIENTS WITH ONCOLOGICAL DISEASES-DATA ON OUTCOMES AND EFFECTIVENESS

Several available COVID-19 vaccines are now in use all over the world. Moderate or severely immunocompromised people should receive a vaccination to protect them from severe COVID-19 disease[60,61].

The efficacy of COVID-19 vaccines in cancer patients is a question of continuous research, with most studies using immunological parameters as surrogate endpoints for clinical outcomes. Clinical trials investigating immune response after COVID-19 vaccination often use seroconversion to SARS-CoV-2 spike (S) protein as an endpoint for vaccine efficacy. Other parameters such as anti-spike antibody titers, detection of neutralizing antibodies, and cellular immune response are usually explored as secondary endpoints[62]. Some authors, however, underscore the role of neutralizing antibodies as the immunological parameter, which probably best correlates with the level of protection after COVID-19 vaccination[63-65].

Both humoral and cellular immune responses to COVID-19 vaccines differ in patients with malignancies compared to noncancer patients; this is not only attributed to the immunosuppressive nature of the oncologic disease but also to the antitumor therapy itself and its direct impact on immune cells. While patients with solid tumors have seroconversion rates similar to the general population, the most significant concern regarding post-vaccination and post-infectious COVID-19 immunity lies with hematological malignancies, especially those where lymphocyte-depletion therapy is used. In support of this is the research of Monin et al[66], who presented interim results of a prospective observational study that explores the immunogenicity of one compared to receiving two doses of the COVID-19 vaccine in patients with cancer by assessing the humoral immune response between 151 patients (95 with solid tumors and 56 with hematological malignancies) and 54 healthy controls. Authors reported efficacy after the first dose in 94%, 38%, and 18% of control subjects, patients with solid tumors and hematological cancer, respectively. After the second dose, the response increased to 100% in controls, 95% in patients with solid cancers, and only 60% in the group with hematological malignancies[66].

When considering post-vaccination immunity in patients with cancer, we should consider that those with hematological malignancies are expected to show different levels of antibody response to COVID-19 vaccines compared to patients with solid tumors. One of the most substantial pieces of evidence in corroboration came from the CAPTURE trial[67]. This prospective clinical study assessed the humoral response after COVID-19 vaccination in more than 700 subjects with solid tumors or hematologic neoplasms, 585 of whom did not have previous SARS-CoV-2 infection. The trial demonstrated 85% and 54% seroconversion rates for anti-spike antibodies after the second dose in patients with solid tumors and hematological malignancies, respectively. However, the response observed among participants was not the same for all SARS-CoV-2 variants[68].

The authors announced substantial differences in neutralizing antibodies concerning viral genotypes from the CAPTURE trial: 83% of patients developed detectable levels of the original SARS-CoV-2 and only 54% of the delta variant. And while nearly two-thirds (62%) of patients with solid tumors elicit humoral response against delta variant, only 31% of those with hematologic malignancies did so[67]. The prospective cohort study of immune response to COVID-19 vaccination in cancer patients CAPTURE (NCT03226886) also showed that among 585 patients, the antibody rates after two doses of BNT162b2 or AZD1222 vaccines given over 12 wk were assessed. The results showed that seroconversion was 85% and 59% after two doses in patients with solid and hematological malignancies, respectively. Neutralizing antibodies against SARS-CoV-2 VOCs were detected in a small proportion of patients, mainly with solid cancers. Vaccine-induced T-cell responses were found in 80% of patients regardless of the vaccine or type of cancer[67].

In an attempt to overcome this relatively low rate of seroconversion in patients with blood cancers, Greenberger et al[69] conducted a large prospective cohort trial on nearly 700 patients vaccinated with three doses of the COVID-19 vaccine. It was estimated that antibody response indeed increased with the 3rd (booster) dose, so 43% of those without detectable antibodies after the 2nd dose demonstrated humoral response after the booster. However, about 20% of all hematological patients still failed to achieve a response even with 3 doses of vaccine[69]. In contrast to the plethora of research on humoral immunity after COVID-19 vaccination in cancer patients, the cellular immune response in this setting is considerably less studied. In a review article by Rüthrich et al[70], the authors tried to summarize what is currently known about the issue in patients with solid tumors and hematological malignancies, comparing data from COVID-19 vaccines and other ”classical” vaccines. Although the assessment of T-cell immune response in the reviewed studies varied, most research used methods based on quantifying and characterizing pathogen-specific T cells and/or estimating T-cell function by cytokine measurement[70].

Observations on immune responses in patients with hematological malignancies revealed that although this population may lack adequate levels of neutralizing viral antibodies, especially after treatment with B cell-depleting agents such as anti-cluster of differentiation 20 monoclonal antibodies, COVID-19 vaccines are still able to produce protective cellular immunity. Solid evidence for the sufficient efficacy of T-cell response comes from a trial in patients with agammaglobulinemia who demonstrated improved COVID-19 infection outcomes after vaccination. However, cellular immunity could also be impaired in this specific patient population, and some of the significant factors for this are age, disease activity, immunosuppressive treatment, and low lymphocyte counts in circulation[70].

This discordance between humoral and cellular immune response could also be seen In patients with solid tumors. In this population, T-cell responses vary among different cancer subtypes and are determined mainly by the type of systemic antitumor treatment. Various studies have demonstrated wide ranges in terms of cellular immunity achieved after COVID-19 vaccination ranging from about 50% to nearly 90% of the vaccinated cases[71,72].

However, despite being generally higher than those observed in blood cancer patients, T-cell response in those with solid tumors remains significantly lower than in healthy controls. One of the most extensive trials reporting data on immune response in patients with solid tumors receiving systemic anticancer treatment is the VOICE study[73]. After recruiting nearly 800 subjects (240 without cancer), the authors assessed cellular immunity by measuring the SARS-CoV-2 spike-specific interferon gamma T-cell response after two vaccine doses. They reported cellular responses in 67%, 66%, and 53% of patients treated with chemotherapy, immunotherapy, or chemoimmunotherapy, respectively. Another interesting trial finding was that more than 40% of patients who did not elicit a humoral immune response could develop a T-cell response, highlighting the vaccine’s ‘double-edge sword’ efficacy in this specific population. Similar to the model observed with the humoral response, whether the cellular response is affected by a booster dose is still an open question since there are conflicting data. Some studies have reported significant enhancement of the T-cell response after the 3rd dose, whereas others refute such assertions[73].

To date, most trials reporting COVID-19 vaccine efficacy in cancer rely on immunological endpoints and not so much on clinical outcomes. However, a recent study on infection rate and outcomes in vaccinated patients with solid tumors and hematologic malignancies has raised concern that despite vaccination, these patients remain at risk of worse outcomes compared to the general population[74]. Among fully vaccinated cancer patients, who experienced breakthrough SARS-CoV-2 infection, the hospitalization rate, intensive care unit admission (or required mechanical vaccination), and death rate are 65%, 19%, and 13%, respectively. This is mainly attributed to patients’ comorbidities and the much worse COVID-19 prognosis in those with hematological malignancies.

In a prospective study conducted by Goshen-Lago et al[75], it was shown that patients with solid tumors demonstrated short-term efficacy and safety of the BNT162b2 vaccine. A follow-up study evaluated these outcomes at 6 mo after vaccination[76]. Participants were 154 patients with solid tumors and 135 controls (health workers). At 6 mo after vaccination, 122 patients were seropositive compared with 114 controls, and the serologic titers dramatically decreased almost equally in both cohorts. Efficacy and safety evidence of BNT162b2 vaccines shows that the serological profile in cancer patients after 6 mo resembles that of the general population[76].

A similar study was conducted by Barrière et al[77], who evaluated the immunogenicity of the BNT162b2 vaccine in patients with solid tumors. Serological analyses were performed during the first vaccination, during the booster dose (w3-w4), and 3-4 wk after the booster dose (w6-w8). The study reported the results for 122 of 194 evaluable patients with solid tumors who had at least two doses from January 2021 to March 2021. In the first analysis (w3-w4), 58 patients had neutralizing antibodies, although the median levels were significantly lower than in the control group. In the following analysis (w6-w8), the data showed the same anti-S seroconversion rate, demonstrating impaired immunogenicity of the BNT162b2 vaccine in cancer patients[77].

Shroff et al[78] also compared anti-S seroconversion to the BNT162b2 mRNA vaccine in patients with solid tumors on active cytotoxic anticancer therapy with healthy control participants. Neutralizing antibodies were found in 67% of cancer patients after the first immunization, and a follow-up analysis found a threefold increase in titers after the second or third doses. European Union Drug Regulating Authorities Clinical Trials (EudraCT) Number 2021-000291-11 was conducted in patients with solid cancers, multiple myeloma, and inflammatory bowel disease[79]. The study is a prospective, open-label, phase four trial to monitor vaccine-specific antibody and cellular responses after booster vaccination with mRNA-1273 or BNT162b2. The data show that booster vaccination against SARS-CoV-2 reverses the lack of response and early antibody weakening in immunocompromised patients.

Another study on the efficacy and safety of heterologous booster vaccination with Ad26.COV2.S after BNT162b2 mRNA vaccine in cancer patients without antibody response was conducted in 2022[80]. The assessment was done directly before vaccination and 4 wk after. Ad26.COV2.S booster vaccination resulted in a serological response in 31% of nonresponders after a double dose of BNT162b2. Clinical trials with the number NCT04368728 reported results from individuals with a history of past or active neoplasms and up to 6 mo of follow-up after dose 2 of a placebo-controlled, observer-blinded trial of the BNT162b2 vaccine[81]. In participants with past or active neoplasms, two doses of the BNT162b2 vaccine improved efficacy and safety profile as in the overall trial population. No vaccine-related deaths were reported.

One of the first evaluations of the effectiveness of vaccination against breakthrough SARS-CoV-2 infections in cancer patients at a population level was done by Lee et al[82]. Analysis was performed in the cancer cohort by vaccine type (BNT162b2, ChAdOx1 nCov-19, or mixed, and other), cancer type and subtype, stage, date of cancer diagnosis, and anticancer treatment or radiotherapy. Data show that vaccination with different COVID-19 vaccines is effective in people with cancer, providing varying levels of protection against SARS-CoV-2 infection. However, it is lower in cancer patients than in the general population[82].

A single-arm prospective clinical trial was conducted with 106 cancer patients by Thakkar et al[83]. They received two doses of mRNA followed by one dose of AD26.CoV2.S vaccine or a third dose of mRNA vaccine. The results showed that a third dose induced immunity in cancer patients. Seroconversion was also assessed in 57% of patients who did not respond to primary vaccination. A fourth dose boosted the immune response by two-thirds. Some patients have neutralizing activity against the omicron variant[83].

In conclusion, all of these studies confirm that people with cancer are at increased risk of severe COVID-19 disease, hospitalization, and death after SARS-CoV-2 infection compared to the general population. The above data show that cancer patients have impaired overall vaccine effectiveness to the approved COVID-19 vaccines. Seroconversion in them decreases faster than in the control population. Although vaccination provides different levels of protection, there should be a global prioritization of the programs to boost vaccination for cancer people, considering the impact of other treatments.

There are still a lack of data on vaccine efficacy in cancer patients concerning novel virus variants like omicron[68]. Table 1 presents the studies on the effectiveness and safety of COVID-19 vaccination with different approved COVID-19 vaccines in oncological patients with solid tumors[5,63,66,67,69-71,73,75-84,84].

Table 1 Some of the more significant studies conducted on the efficacy and safety of COVID-19 vaccination with different approved COVID-19 vaccines in oncological patients with solid tumors.
Ref.
Type of vaccine
Type of study
Subjects (diagnosis, other specific characteristics)
Data on efficacy
Data on safety (main side effects)
OnCovid study group[5]NAA multicenter observational registry-based studyAll PTs included n = 2634 (100%); PTs with advanced tumor stage n = 1244 (46%); PTs with receipt of anti-cancer therapy within 4 wk of COVID-19 diagnosis n = 1305 (51.8%); malignancy type: Breast n = 493 (18.9%); gastrointestinal n = 476 (18.2%); gynecologic/genitourinary n = 530 (20.3%); hematologic n = 357 (13.7%)The difference in the necessity of hospitalization due to COVID-19, oxygen therapy requirement, mechanical ventilation requirement, and 14-d CFR between PTs stratified across time five phases and two major outbreaks of the pandemic; hospitalization requirement: 1st phase-64.7% to 5th phase-42.7% (P < 0.01); proportion to PTs requiring oxygen: therapy, phase 1-62.6%, to phase 5-46.0% (P < 0.001); mechanical ventilation: Phase 1-12.1% to phase 5-11.8% (P = 0.01); CFR: 1st outbreak-25.6% to-2nd outbreak 16.2% (P < 0.001)N/A
Khoury et al[63], 2021mRNA and adenoviral vector vaccines20.2% of subjects had (95%CI) 50% protective neutralization levelN/A
Monin et al[66], 2021mRNAProspective observational studyPTs with oncologic disease n = 151: With solid cancer n = 95; with hematological malignancy n = 56; and HCs n = 54Surrogate marker of efficiency: Seroconversion after 1st dose: 32 of 34 (94%) HCs, 21 of 56 (38%), PTs with solid cancer, 8 of 44 (18%) PTs with hematologic malignancies; after 2nd dose: 12 of 12 (100%) HCs; 18 of 19 (95%) PTs with solid, 3 of 5 (60%) PTs with hematologic malignanciesAE: Injection site pain within 7 d following the first dose in: 23 of 65 (35%) PTs with cancer; 12 of 25 (48%) HCs; no vaccine-related deaths were reported
Greenberger et al[69], 2021mRNA and adenoviral vector vaccinesRetrospective cohort studyPTs with hematologic malignancies, n = 3300
Ehmsen et al[71], 2021mRNAProspective cohort study (comparison between groups with different malignancies; no HCs)PTs with cancer, n = 524, of whom: 201 (38%) with solid cancer; 323 (62%) with hematologic cancer; 524 (100%) had a blood sample drawn at a median of 36 d after the second dose of vaccine; and 247 (47%) had a second blood sample drawn 3 mo after the second dose of the vaccineSeropositivity rate for anti-S IgG 36 d after vaccination: PTs with solid cancer 187 of 201 (93%); PTs with hematologic cancer 215 of 323 (66%); seropositivity rate for anti-S IgG 3 mo after vaccination: PTs with solid cancer-86%, PTs with hematologic cancer-53%; anti-S IgG titers; between 36-d and 3-mo samples declined from a median of 429 BAU/mL to a median of 139 BAU/mL (P = 0.03, Student’s t-test); T-cell reactivity: PTs with solid cancer-92 (46%), 70 (76%) mounted both CD4+ and CD8+ T-cell responses, 21 (23%) elicited only a CD8+ T-cell response, PTs with hematologic cancer-144 (45%), 81% were positive for both CD4+ and CD8+ T cells, 26 (18%) only elicited a CD8+ T cell response, 76% of the seronegative PTs did not elicit a T-cell response; PTs with solid cancer: only 1 of the 14 (7%) seronegative PTs elicited a T-cell response; PTs with hematologic cancer: 28 of 108 (26%) PTs elicited a T-cell responseN/A
Oosting et al[73], 2021mRNAProspective, multicenter, non-inferiority trialCohort A: Individuals without cancer (control cohort); cohort B: PTs with SOTs, regardless of stage and histology, treated with immunotherapy; cohort C: PTs treated with chemotherapy; and cohort D: PTs treated with chemoimmunotherapyPresence of SARS-CoV-2-binding antibodies after the second vaccination; at 28th d, 6 mo after 12 mo after a spike-specific T-cell response was defined as a two times or more significant increase in the number of spot-forming cellsN/A
Polack et al[84], 2020mRNA vaccinesPlacebo-controlled, observer-blinded, pivotal efficacy trial (randomized 1:1 vaccine vs placebo)All PTs included n = 43548; PTs with liver disease n = 217 (0.6%)95% efficacy (9 vaccinated vs 169 controls with COVID-19); 10 cases of severe COVID-19 infection vs 9 in the placebo group; flares: NRSystemic AEs: (1) Fatigue (34%-51%); (2) headache (25%-39%); (3) fever (11%), injection site reactions; (4) pain (71%-83%); (5) redness and swelling (< 7%); and (6) serious AE < 4%
Fendler et al[67], 2021BNT162b2 or AZD1222 vaccines (CAPTURE, NCT03226886)Prospective cohort study585 PTs, the seroconversion rates after two doses of BNT162b2 or AZD1222 vaccines given over 12 wk were assessedAfter two doses of BNT162b2 or AZD1222 vaccines given over 12 wk, seroconversion was 85% and 59% in PTs with solid and hematological malignancies, respectively; vaccine-induced T-cell responses were found in 80% of PTs regardless of the vaccine or type of cancerN/A
Goshen-Lago et al[75], 2021BNT162b2 vaccineProspective study154 PTs with SOTs and 135 HCs (health workers)In PTs with cancer with active intravenous treatment, 79% (n = 122) of the PTs had positive serologic test results, compared with 84% (n = 114) in the control group; analysis by age, sex, or disease stage has no significant differences within the PT cohort; 15% of the seropositive PTs became seronegative after 6 mo, comparable to the control groupN/A
Waldhorn et al[76], 2021BNT162b2 vaccineProspective study154 PTs with SOTs and 135 controls6 mo postvaccination, 79% of PTs and 84% of HCs were seropositive (P = 0.32); dramatically decreased serology titer
Shroff et al[78], 2021BNT162b2Phase 1 cohort trial53 PTs with SOTs on active cytotoxic anticancer therapy and 50 healthy cohortNeutralizing antibodies were detected in 67% of PTs with cancer after the first immunization, followed by a threefold increase in median titers after the second doseAEs were mild: temperature, fever, headache, redness, and swelling on the injection site
Barrière et al[77], 2021BNT162b2VMO for vaccinated PTs under active treatment in the Department of Oncology of the Saint Jean Polyclinic, Nice, France194 evaluable PTs with SOTs and 31 HCs58 PTs had neutralizing antibodies, although the median levels were significantly lower than those in the control group; the data demonstrating impaired immunogenicity of the BNT162b2 vaccine in immunocompromised PTs; % of efficacy was not reportedN/A
Thomas et al[81], 2022BNT162b2 mRNAPhase 3 randomized clinical trial3813 participants had a history of neoplasm: Most common malignancies were breast (n = 460), prostate (n = 362), and melanoma (n = 223)Vaccine efficacy was 94.4% (95%CI) after up to 6 mo of follow-up post-dose 2N/A
Wagner et al[79], 2022mRNA-1273 or BNT162b2Prospective, open-label, phase four trail263 PTs with SOT, n = 63), MM, n = 70, IBD, n = 130 and 66 controls1 mo after the two-dose primary vaccination, the highest nonresponder rate was found in MM PTs (17%); 6 mo after the second dose, 18% of PTs with MM, 10% with SOT, and 4% with IBD became seronegative compared to the control group; the vaccination with mRNA-1273 led to higher antibody levels than with BNT162b2; booster vaccination increased antibody levels 8-fold in seropositive individuals and induced responses in those with undetectable pre-booster antibody levelsN/A
Lee et al[82], 2022BNT162b2, ChAdOx1 nCov-19, or mixed and otherPopulation-based test-negative case-control studyCancer cohort comprised 377194 individuals, of whom 42882 had breakthrough SARS-CoV-2 infections; the control population consisted of 28010955 individuals, of whom 5748708 had SARS-CoV-2 breakthrough infectionsOverall vaccine effectiveness was 69.8% in the control population and 65.5% in the cancer cohort; vaccine effectiveness at 3-6 mo was lower in the cancer cohort (47.0%) than in the control population (61.4%)N/A
Reimann et al[80], 2022Ad26.COV2.S after BNT162b2 mRNA32 oncological nonresponders to double-dose BNT162b2The overall response rate was 31%Mainly mild local and systemic reactions
Thakkar et al[83], 2023Two doses of mRNA or one dose of AD26.CoV2.S vaccine and administered a third dose of mRNA vaccineSingle-arm prospective clinical trialcancer PTsA third dose of the COVID-19 vaccine induces durable immunity in cancer PTs, leading to seroconversion in 57% of PTs who did not respond to primary vaccination; 18 PTs with blood cancer and severe immune suppression had no response after three doses; and the fourth dose boosted the immune response by 2/3 of PTs, with neutralizing activity against the omicron variantN/A
COVID-19 VACCINES AND CHEMOTHERAPY INTERACTIONS

People with cancer often have an increased susceptibility to infections due to various factors, including cancer itself and/or, in some cases, the applied therapy, poor nutrition, and damaged physiological barriers. In addition, the incidence of neoplasia is highest in individuals aged 65 and over. When the immune system’s effectiveness is weakened, the elderly often have concomitant diseases for which they can also take medications[54,85].

Regarding cancer chemotherapy, conventional antitumor chemotherapeutic agents kill actively proliferating cells, including bone marrow cells, and myelosuppression is one of clinical oncology’s most common side effects[86]. Chemotherapy-induced neutropenia is a significant cause of hematological and dose-limiting toxicities of chemotherapy[87]. Some currently available anticancer drugs, such as methotrexate and cyclophosphamide, express immunosuppressive effects and impair peripheral T cells’ proliferative and/or effector functions. Methotrexate is an antimetabolite of the antifolate type developed in 1947 and is included in the World Health Organization’s List of Essential Medicines. Currently, it is widely used not only in clinical oncology (in the treatment of acute lymphoblastic leukemia, acute myeloid leukemia, meningeal leukemia and lymphoma, osteosarcomas, non-Hodgkin’s lymphoma, breast and bladder cancers, etc.) but also as a first-line treatment in autoimmune, inflammatory diseases such as rheumatoid arthritis, psoriasis and Crone’s disease[88-90]. Methotrexate has been found to disturb antibody response after pneumococcal vaccination[91,92]; the drug reduces circulating T helper 17 (Th17) cells and impairs plasmablast and memory B-cell expansion following pneumococcal conjugate immunization in patients with rheumatoid arthritis[93].

Cyclophosphamide is an alkylating agent synthesized in 1958 and used for decades in clinical practice in the therapy regimens of neoplasms (malignant lymphomas, multiple myeloma, sarcoma, breast cancer, disseminated neuroblastomas, retinoblastoma, ovarian adenocarcinoma, etc) and as an immunosuppressive agent for the treatment of autoimmune and immune-mediated diseases such as multiple sclerosis. Cyclophosphamide shows selectivity for T cells and is an immunosuppressant to prevent transplant rejection and graft-vs-host complications[94]. Cyclophosphamide has been associated with suppressing helper Th1 activity and enhancing Th2 responses[95]. This drug inhibits Th1/Th17 responses and increases the cells secreting anti-inflammatory cytokines such as IL-4, IL-10, and transforming growth factor beta[96]. A single administration of low-dose cyclophosphamide selectively suppresses regulatory T cells. The low-dose cyclophosphamide promotes antitumor immunity by selectively depleting regulatory T cells and enhancing effector T cell function. However, cyclophosphamide can also increase the number of myeloid-derived suppressor cells[97,98].

Treatment with tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib applied in the treatment of chronic myeloid leukemia is associated with loss of memory B-cell subsets and impaired humoral immune responses to 23-valent polysaccharide pneumococcal vaccine, likely due to the off-target kinase inhibitory activity of these drugs[99].

CONCLUSION

Data so far show that patients with cancer are at increased risk of severe COVID-19 and developing various complications mainly due to their immunocompromised state, type of treatment and comorbidities. Although cancer patients were excluded from vaccine trials, the investigations of post-vaccinal immune responses and the effectiveness of the vaccines showed that both humoral and cellular immune responses to COVID-19 vaccines differ in patients with malignancies compared to noncancer patients, and this is being attributed not only to the immunosuppressive nature of the oncologic disease but to the antitumor therapy itself and its direct impact on immune cells.

The evidence indicates that the efficacy of vaccinations could be impaired in cancer patients in line with a reduced rate of seroconversion and shorter duration compared to healthy controls. Despite these data, when focusing on the clinical outcomes instead of immunological endpoints regarding vaccine efficacy, COVID-19 vaccines demonstrated high effectiveness in preventing severe COVID-19 and infection-related death, and safety profile with comparable to healthy controls adverse effects in patients with solid tumors and hematological malignancies.

Despite the considerably higher mortality in the cancer patients group from COVID-19 than the general population, countries with high vaccination rates have demonstrated trends toward improved survival of cancer patients early and late in the pandemic. Nevertheless, vaccination of these patients and overall vaccination of the population has proven to significantly reduce the risk of complications and mortality of COVID-19 and should be promoted worldwide.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country/Territory of origin: Bulgaria

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Chen LJ, China; Emran TB, Bangladesh S-Editor: Chen YL L-Editor: Filipodia P-Editor: Chen YL

References
1.  Yu J, Ouyang W, Chua MLK, Xie C. SARS-CoV-2 Transmission in Patients With Cancer at a Tertiary Care Hospital in Wuhan, China. JAMA Oncol. 2020;6:1108-1110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 626]  [Cited by in F6Publishing: 746]  [Article Influence: 186.5]  [Reference Citation Analysis (0)]
2.  Lewis MA. Between Scylla and Charybdis-Oncologic Decision Making in the Time of Covid-19. N Engl J Med. 2020;382:2285-2287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
3.  ElGohary GM, Hashmi S, Styczynski J, Kharfan-Dabaja MA, Alblooshi RM, de la Cámara R, Mohmed S, Alshaibani A, Cesaro S, Abd El-Aziz N, Almaghrabi R, Gergis U, Majhail NS, El-Gohary Y, Chemaly RF, Aljurf M, El Fakih R. The Risk and Prognosis of COVID-19 Infection in Cancer Patients: A Systematic Review and Meta-Analysis. Hematol Oncol Stem Cell Ther. 2022;15:45-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 74]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
4.  Vijenthira A, Gong IY, Fox TA, Booth S, Cook G, Fattizzo B, Martín-Moro F, Razanamahery J, Riches JC, Zwicker J, Patell R, Vekemans MC, Scarfò L, Chatzikonstantinou T, Yildiz H, Lattenist R, Mantzaris I, Wood WA, Hicks LK. Outcomes of patients with hematologic malignancies and COVID-19: a systematic review and meta-analysis of 3377 patients. Blood. 2020;136:2881-2892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 436]  [Article Influence: 145.3]  [Reference Citation Analysis (0)]
5.  OnCovid Study Group; Pinato DJ, Patel M, Scotti L, Colomba E, Dolly S, Loizidou A, Chester J, Mukherjee U, Zambelli A, Dalla Pria A, Aguilar-Company J, Bower M, Salazar R, Bertuzzi A, Brunet J, Lambertini M, Tagliamento M, Pous A, Sita-Lumsden A, Srikandarajah K, Colomba J, Pommeret F, Seguí E, Generali D, Grisanti S, Pedrazzoli P, Rizzo G, Libertini M, Moss C, Evans JS, Russell B, Harbeck N, Vincenzi B, Biello F, Bertulli R, Ottaviani D, Liñan R, Rossi S, Carmona-García MC, Tondini C, Fox L, Baggi A, Fotia V, Parisi A, Porzio G, Queirolo P, Cruz CA, Saoudi-Gonzalez N, Felip E, Roqué Lloveras A, Newsom-Davis T, Sharkey R, Roldán E, Reyes R, Zoratto F, Earnshaw I, Ferrante D, Marco-Hernández J, Ruiz-Camps I, Gaidano G, Patriarca A, Bruna R, Sureda A, Martinez-Vila C, Sanchez de Torre A, Berardi R, Giusti R, Mazzoni F, Guida A, Rimassa L, Chiudinelli L, Franchi M, Krengli M, Santoro A, Prat A, Tabernero J, Van Hemelrijck M, Diamantis N, Gennari A, Cortellini A. Time-Dependent COVID-19 Mortality in Patients With Cancer: An Updated Analysis of the OnCovid Registry. JAMA Oncol. 2022;8:114-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 46]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
6.  Chen L, Qu J, Kalyani FS, Zhang Q, Fan L, Fang Y, Li Y, Xiang C. Mesenchymal stem cell-based treatments for COVID-19: status and future perspectives for clinical applications. Cell Mol Life Sci. 2022;79:142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
7.  Yuan Y, Jiao B, Qu L, Yang D, Liu R. The development of COVID-19 treatment. Front Immunol. 2023;14:1125246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 50]  [Reference Citation Analysis (0)]
8.  Gasparyan AY, Ayvazyan L, Blackmore H, Kitas GD. Writing a narrative biomedical review: considerations for authors, peer reviewers, and editors. Rheumatol Int. 2011;31:1409-1417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 436]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
9.  Li H, Baldwin E, Zhang X, Kenost C, Luo W, Calhoun EA, An L, Bennett CL, Lussier YA. Comparison and impact of COVID-19 for patients with cancer: a survival analysis of fatality rate controlling for age, sex and cancer type. BMJ Health Care Inform. 2021;28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  He W, Chen L, Yuan G, Fang Y, Chen W, Wu D, Liang B, Lu X, Ma Y, Li L, Wang H, Chen Z, Li Q, Gale RP. COVID-19 in persons with haematological cancers. Leukemia. 2020;34:1637-1645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 323]  [Article Influence: 80.8]  [Reference Citation Analysis (0)]
11.  Zhang H, Han H, He T, Labbe KE, Hernandez AV, Chen H, Velcheti V, Stebbing J, Wong KK. Clinical Characteristics and Outcomes of COVID-19-Infected Cancer Patients: A Systematic Review and Meta-Analysis. J Natl Cancer Inst. 2021;113:371-380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 121]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
12.  Aries JA, Davies JK, Auer RL, Hallam SL, Montoto S, Smith M, Sevillano B, Foggo V, Wrench B, Zegocki K, Agrawal S, Le Dieu R, Truelove E, Erblich T, Araf S, Okosun J, Oakervee H, Cavenagh JD, Gribben JG, Riches JC. Clinical outcome of coronavirus disease 2019 in haemato-oncology patients. Br J Haematol. 2020;190:e64-e67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
13.  Tsamakis K, Gavriatopoulou M, Schizas D, Stravodimou A, Mougkou A, Tsiptsios D, Sioulas V, Spartalis E, Sioulas AD, Tsamakis C, Charalampakis N, Mueller C, Arya D, Zarogoulidis P, Spandidos DA, Dimopoulos MA, Papageorgiou C, Rizos E. Oncology during the COVID-19 pandemic: challenges, dilemmas and the psychosocial impact on cancer patients. Oncol Lett. 2020;20:441-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
14.  Iloanusi S, Mgbere O, Essien EJ. Polypharmacy among COVID-19 patients: A systematic review. J Am Pharm Assoc (2003). 2021;61:e14-e25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
15.  Yang F, Shi S, Zhu J, Shi J, Dai K, Chen X. Clinical characteristics and outcomes of cancer patients with COVID-19. J Med Virol. 2020;92:2067-2073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 133]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
16.  Afshar ZM, Dayani M, Naderi M, Ghanbarveisi F, Shiri S, Rajati F. Fatality rate of COVID-19 in patients with malignancies: a sytematic review and meta-analysis. J Infect. 2020;81:e114-e116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
17.  Brar G, Pinheiro LC, Shusterman M, Swed B, Reshetnyak E, Soroka O, Chen F, Yamshon S, Vaughn J, Martin P, Paul D, Hidalgo M, Shah MA. COVID-19 Severity and Outcomes in Patients With Cancer: A Matched Cohort Study. J Clin Oncol. 2020;38:3914-3924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 95]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
18.  Onder G, Rezza G, Brusaferro S. Case-Fatality Rate and Characteristics of Patients Dying in Relation to COVID-19 in Italy. JAMA. 2020;323:1775-1776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1653]  [Cited by in F6Publishing: 2089]  [Article Influence: 522.3]  [Reference Citation Analysis (0)]
19.  Guven DC, Aktas BY, Aksun MS, Ucgul E, Sahin TK, Yildirim HC, Guner G, Kertmen N, Dizdar O, Kilickap S, Aksoy S, Yalcin S, Turker A, Uckun FM, Arik Z. COVID-19 pandemic: changes in cancer admissions. BMJ Support Palliat Care. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
20.  Skovlund CW, Friis S, Christensen J, Nilbert MC, Mørch LS. Drop in cancer diagnosis during the COVID-19 pandemic in Denmark: assessment of impact during 2020. Acta Oncol. 2022;61:658-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
21.  Vázquez Rosas T, Cazap E, Delgado L, Ismael J, Bejarano S, Castro C, Castro H, Müller B, Gutiérrez-Delgado F, Santini LA, Vallejos Sologuren C. Social Distancing and Economic Crisis During COVID-19 Pandemic Reduced Cancer Control in Latin America and Will Result in Increased Late-Stage Diagnoses and Expense. JCO Glob Oncol. 2021;7:694-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
22.  Yong JH, Mainprize JG, Yaffe MJ, Ruan Y, Poirier AE, Coldman A, Nadeau C, Iragorri N, Hilsden RJ, Brenner DR. The impact of episodic screening interruption: COVID-19 and population-based cancer screening in Canada. J Med Screen. 2021;28:100-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 116]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
23.  Salunke AA, Nandy K, Pathak SK, Shah J, Kamani M, Kottakota V, Thivari P, Pandey A, Patel K, Rathod P, Bhatt S, Dave P, Pandya S. Impact of COVID -19 in cancer patients on severity of disease and fatal outcomes: A systematic review and meta-analysis. Diabetes Metab Syndr. 2020;14:1431-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
24.  Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, Wang B, Xiang H, Cheng Z, Xiong Y, Zhao Y, Li Y, Wang X, Peng Z. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA. 2020;323:1061-1069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14113]  [Cited by in F6Publishing: 14268]  [Article Influence: 3567.0]  [Reference Citation Analysis (0)]
25.  Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, Iotti G, Latronico N, Lorini L, Merler S, Natalini G, Piatti A, Ranieri MV, Scandroglio AM, Storti E, Cecconi M, Pesenti A; COVID-19 Lombardy ICU Network. Baseline Characteristics and Outcomes of 1591 Patients Infected With SARS-CoV-2 Admitted to ICUs of the Lombardy Region, Italy. JAMA. 2020;323:1574-1581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3537]  [Cited by in F6Publishing: 3669]  [Article Influence: 917.3]  [Reference Citation Analysis (0)]
26.  Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW; the Northwell COVID-19 Research Consortium, Barnaby DP, Becker LB, Chelico JD, Cohen SL, Cookingham J, Coppa K, Diefenbach MA, Dominello AJ, Duer-Hefele J, Falzon L, Gitlin J, Hajizadeh N, Harvin TG, Hirschwerk DA, Kim EJ, Kozel ZM, Marrast LM, Mogavero JN, Osorio GA, Qiu M, Zanos TP. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA. 2020;323:2052-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6024]  [Cited by in F6Publishing: 6229]  [Article Influence: 1557.3]  [Reference Citation Analysis (0)]
27.  Kuderer NM, Choueiri TK, Shah DP, Shyr Y, Rubinstein SM, Rivera DR, Shete S, Hsu CY, Desai A, de Lima Lopes G Jr, Grivas P, Painter CA, Peters S, Thompson MA, Bakouny Z, Batist G, Bekaii-Saab T, Bilen MA, Bouganim N, Larroya MB, Castellano D, Del Prete SA, Doroshow DB, Egan PC, Elkrief A, Farmakiotis D, Flora D, Galsky MD, Glover MJ, Griffiths EA, Gulati AP, Gupta S, Hafez N, Halfdanarson TR, Hawley JE, Hsu E, Kasi A, Khaki AR, Lemmon CA, Lewis C, Logan B, Masters T, McKay RR, Mesa RA, Morgans AK, Mulcahy MF, Panagiotou OA, Peddi P, Pennell NA, Reynolds K, Rosen LR, Rosovsky R, Salazar M, Schmidt A, Shah SA, Shaya JA, Steinharter J, Stockerl-Goldstein KE, Subbiah S, Vinh DC, Wehbe FH, Weissmann LB, Wu JT, Wulff-Burchfield E, Xie Z, Yeh A, Yu PP, Zhou AY, Zubiri L, Mishra S, Lyman GH, Rini BI, Warner JL; COVID-19 and Cancer Consortium. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet. 2020;395:1907-1918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1195]  [Cited by in F6Publishing: 1215]  [Article Influence: 303.8]  [Reference Citation Analysis (0)]
28.  Jee J, Foote MB, Lumish M, Stonestrom AJ, Wills B, Narendra V, Avutu V, Murciano-Goroff YR, Chan JE, Derkach A, Philip J, Belenkaya R, Kerpelev M, Maloy M, Watson A, Fong C, Janjigian Y, Diaz LA Jr, Bolton KL, Pessin MS. Chemotherapy and COVID-19 Outcomes in Patients With Cancer. J Clin Oncol. 2020;38:3538-3546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 174]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
29.  Mukkada S, Bhakta N, Chantada GL, Chen Y, Vedaraju Y, Faughnan L, Homsi MR, Muniz-Talavera H, Ranadive R, Metzger M, Friedrich P, Agulnik A, Jeha S, Lam C, Dalvi R, Hessissen L, Moreira DC, Santana VM, Sullivan M, Bouffet E, Caniza MA, Devidas M, Pritchard-Jones K, Rodriguez-Galindo C; Global Registry of COVID-19 in Childhood Cancer. Global characteristics and outcomes of SARS-CoV-2 infection in children and adolescents with cancer (GRCCC): a cohort study. Lancet Oncol. 2021;22:1416-1426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 81]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
30.  Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, Lichtor T, Decker WK, Whelan RL, Kumara HMCS, Signori E, Honoki K, Georgakilas AG, Amin A, Helferich WG, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Keith WN, Bilsland A, Bhakta D, Halicka D, Fujii H, Aquilano K, Ashraf SS, Nowsheen S, Yang X, Choi BK, Kwon BS. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015;35 Suppl:S185-S198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 938]  [Article Influence: 104.2]  [Reference Citation Analysis (0)]
31.  Hiam-Galvez KJ, Allen BM, Spitzer MH. Systemic immunity in cancer. Nat Rev Cancer. 2021;21:345-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 325]  [Cited by in F6Publishing: 590]  [Article Influence: 196.7]  [Reference Citation Analysis (0)]
32.  Nesmiyanov PP. Dendritic Cells. Encyclop of Infect and Immun. 2022;1:110-117.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2:1096-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1375]  [Cited by in F6Publishing: 1343]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
34.  Menetrier-Caux C, Montmain G, Dieu MC, Bain C, Favrot MC, Caux C, Blay JY. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: role of interleukin-6 and macrophage colony-stimulating factor. Blood. 1998;92:4778-4791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 369]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
35.  Kiertscher SM, Luo J, Dubinett SM, Roth MD. Tumors promote altered maturation and early apoptosis of monocyte-derived dendritic cells. J Immunol. 2000;164:1269-1276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 181]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
36.  Pinzon-Charry A, Maxwell T, McGuckin MA, Schmidt C, Furnival C, López JA. Spontaneous apoptosis of blood dendritic cells in patients with breast cancer. Breast Cancer Res. 2006;8:R5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
37.  Della Bella S, Gennaro M, Vaccari M, Ferraris C, Nicola S, Riva A, Clerici M, Greco M, Villa ML. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br J Cancer. 2003;89:1463-1472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 164]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
38.  Mastelic-Gavillet B, Sarivalasis A, Lozano LE, Wyss T, Inoges S, de Vries IJM, Dartiguenave F, Jichlinski P, Derrè L, Coukos G, Melero I, Harari A, Romero P, Viganó S, Kandalaft LE. Quantitative and qualitative impairments in dendritic cell subsets of patients with ovarian or prostate cancer. Eur J Cancer. 2020;135:173-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
39.  Tabarkiewicz J, Rybojad P, Jablonka A, Rolinski J. CD1c+ and CD303+ dendritic cells in peripheral blood, lymph nodes and tumor tissue of patients with non-small cell lung cancer. Oncol Rep. 2008;19:237-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
40.  Lu Y, Xu W, Gu Y, Chang X, Wei G, Rong Z, Qin L, Chen X, Zhou F. Non-small Cell Lung Cancer Cells Modulate the Development of Human CD1c(+) Conventional Dendritic Cell Subsets Mediated by CD103 and CD205. Front Immunol. 2019;10:2829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
41.  Legitimo A, Consolini R, Failli A, Orsini G, Spisni R. Dendritic cell defects in the colorectal cancer. Hum Vaccin Immunother. 2014;10:3224-3235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 63]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
42.  Failli A, Legitimo A, Orsini G, Romanini A, Consolini R. Numerical defect of circulating dendritic cell subsets and defective dendritic cell generation from monocytes of patients with advanced melanoma. Cancer Lett. 2013;337:184-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
43.  Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2369]  [Cited by in F6Publishing: 2692]  [Article Influence: 224.3]  [Reference Citation Analysis (0)]
44.  Wang WW, Yuan XL, Chen H, Xie GH, Ma YH, Zheng YX, Zhou YL, Shen LS. CD19+CD24hiCD38hiBregs involved in downregulate helper T cells and upregulate regulatory T cells in gastric cancer. Oncotarget. 2015;6:33486-33499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 101]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
45.  Ray-Coquard I, Cropet C, Van Glabbeke M, Sebban C, Le Cesne A, Judson I, Tredan O, Verweij J, Biron P, Labidi I, Guastalla JP, Bachelot T, Perol D, Chabaud S, Hogendoorn PC, Cassier P, Dufresne A, Blay JY; European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009;69:5383-5391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 443]  [Cited by in F6Publishing: 553]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
46.  Wang JL, Ma R, Kong W, Zhao R, Wang YY. Lymphopenia in Esophageal Cancer: What Have We Learned? Front Oncol. 2021;11:625963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
47.  Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020;10:727-742.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Ye Q, Wang C, Xian J, Zhang M, Cao Y. Expression of programmed cell death protein 1 (PD-1) and indoleamine 2,3-dioxygenase (IDO) in the tumor microenvironment and in tumor-draining lymph nodes of breast cancer. Hum Pathol. 2018;75:81-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 17]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
49.  Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck-Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res. 2003;9:606-612.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Liu L, Wu G, Yao JX, Ding Q, Huang SA. CD4+CD25high regulatory cells in peripheral blood of cancer patients. Neuro Endocrinol Lett. 2008;29:240-245.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Sarvaria A, Madrigal JA, Saudemont A. B cell regulation in cancer and anti-tumor immunity. Cell Mol Immunol. 2017;14:662-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 397]  [Article Influence: 56.7]  [Reference Citation Analysis (0)]
52.  Murakami Y, Saito H, Shimizu S, Kono Y, Shishido Y, Miyatani K, Matsunaga T, Fukumoto Y, Ashida K, Sakabe T, Nakayama Y, Fujiwara Y. Increased regulatory B cells are involved in immune evasion in patients with gastric cancer. Sci Rep. 2019;9:13083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
53.  Li JH, O'Sullivan TE. Back to the Future: Spatiotemporal Determinants of NK Cell Antitumor Function. Front Immunol. 2021;12:816658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
54.  Zembower TR. Epidemiology of infections in cancer patients. Cancer Treat Res. 2014;161:43-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
55.  Bitterman R, Eliakim-Raz N, Vinograd I, Zalmanovici Trestioreanu A, Leibovici L, Paul M. Influenza vaccines in immunosuppressed adults with cancer. Cochrane Database Syst Rev. 2018;2:CD008983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
56.  Brydak LB, Guzy J, Starzyk J, Machała M, Góźdź SS. Humoral immune response after vaccination against influenza in patients with breast cancer. Support Care Cancer. 2001;9:65-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
57.  Pollyea DA, Brown JM, Horning SJ. Utility of influenza vaccination for oncology patients. J Clin Oncol. 2010;28:2481-2490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
58.  Oates HF, Stoker LM, Monaghan JC, Stokes GS. The beta-adrenoceptor controlling renin release. Arch Int Pharmacodyn Ther. 1978;234:205-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
59.  Arrowood JR, Hayney MS. Immunization recommendations for adults with cancer. Ann Pharmacother. 2002;36:1219-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
60.  Velikova T, Georgiev T. SARS-CoV-2 vaccines and autoimmune diseases amidst the COVID-19 crisis. Rheumatol Int. 2021;41:509-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 102]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
61.  Sen P, Ravichandran N, Nune A, Lilleker JB, Agarwal V, Kardes S, Kim M, Day J, Milchert M, Gheita T, Salim B, Velikova T, Gracia-Ramos AE, Parodis I, Selva O'Callaghan A, Nikiphorou E, Chatterjee T, Tan AL, Cavagna L, Saavedra MA, Shinjo SK, Ziade N, Knitza J, Kuwana M, Distler O, Chinoy H, Aggarwal R, Gupta L; COVAD Study Group. COVID-19 vaccination-related adverse events among autoimmune disease patients: results from the COVAD study. Rheumatology (Oxford). 2022;62:65-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
62.  Corti C, Antonarelli G, Scotté F, Spano JP, Barrière J, Michot JM, André F, Curigliano G. Seroconversion rate after vaccination against COVID-19 in patients with cancer-a systematic review. Ann Oncol. 2022;33:158-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 58]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
63.  Khoury DS, Cromer D, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, Subbarao K, Kent SJ, Triccas JA, Davenport MP. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27:1205-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2207]  [Cited by in F6Publishing: 2472]  [Article Influence: 824.0]  [Reference Citation Analysis (0)]
64.  Earle KA, Ambrosino DM, Fiore-Gartland A, Goldblatt D, Gilbert PB, Siber GR, Dull P, Plotkin SA. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021;39:4423-4428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 592]  [Article Influence: 197.3]  [Reference Citation Analysis (0)]
65.  Slabakova Y, Gerenska D, Ivanov N, Velikova T. Immune titers of protection against severe acute respiratory syndrome coronavirus 2: are we there yet? Explor Immunol. 2022;2:9-24.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Monin L, Laing AG, Muñoz-Ruiz M, McKenzie DR, Del Molino Del Barrio I, Alaguthurai T, Domingo-Vila C, Hayday TS, Graham C, Seow J, Abdul-Jawad S, Kamdar S, Harvey-Jones E, Graham R, Cooper J, Khan M, Vidler J, Kakkassery H, Sinha S, Davis R, Dupont L, Francos Quijorna I, O'Brien-Gore C, Lee PL, Eum J, Conde Poole M, Joseph M, Davies D, Wu Y, Swampillai A, North BV, Montes A, Harries M, Rigg A, Spicer J, Malim MH, Fields P, Patten P, Di Rosa F, Papa S, Tree T, Doores KJ, Hayday AC, Irshad S. Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study. Lancet Oncol. 2021;22:765-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 413]  [Article Influence: 137.7]  [Reference Citation Analysis (0)]
67.  Fendler A, Shepherd STC, Au L, Wilkinson KA, Wu M, Byrne F, Cerrone M, Schmitt AM, Joharatnam-Hogan N, Shum B, Tippu Z, Rzeniewicz K, Boos LA, Harvey R, Carlyle E, Edmonds K, Del Rosario L, Sarker S, Lingard K, Mangwende M, Holt L, Ahmod H, Korteweg J, Foley T, Bazin J, Gordon W, Barber T, Emslie-Henry A, Xie W, Gerard CL, Deng D, Wall EC, Agua-Doce A, Namjou S, Caidan S, Gavrielides M, MacRae JI, Kelly G, Peat K, Kelly D, Murra A, Kelly K, O'Flaherty M, Dowdie L, Ash N, Gronthoud F, Shea RL, Gardner G, Murray D, Kinnaird F, Cui W, Pascual J, Rodney S, Mencel J, Curtis O, Stephenson C, Robinson A, Oza B, Farag S, Leslie I, Rogiers A, Iyengar S, Ethell M, Messiou C, Cunningham D, Chau I, Starling N, Turner N, Welsh L, van As N, Jones RL, Droney J, Banerjee S, Tatham KC, O'Brien M, Harrington K, Bhide S, Okines A, Reid A, Young K, Furness AJS, Pickering L, Swanton C; Crick COVID19 consortium, Gandhi S, Gamblin S, Bauer DL, Kassiotis G, Kumar S, Yousaf N, Jhanji S, Nicholson E, Howell M, Walker S, Wilkinson RJ, Larkin J, Turajlic S. Adaptive immunity and neutralizing antibodies against SARS-CoV-2 variants of concern following vaccination in patients with cancer: The CAPTURE study. Nat Cancer. 2021;2:1321-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 95]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
68.  Miteva D, Kitanova M, Batselova H, Lazova S, Chervenkov L, Peshevska-Sekulovska M, Sekulovski M, Gulinac M, Vasilev GV, Tomov L, Velikova T. The End or a New Era of Development of SARS-CoV-2 Virus: Genetic Variants Responsible for Severe COVID-19 and Clinical Efficacy of the Most Commonly Used Vaccines in Clinical Practice. Vaccines (Basel). 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
69.  Greenberger LM, Saltzman LA, Senefeld JW, Johnson PW, DeGennaro LJ, Nichols G. Sars-Cov-2 Antibody Levels in Blood Cancer Patients after a Third Sars-Cov-2 “Booster” Vaccination - Observational Data from the LLS National Registry. Blood. 2021;138 Suppl 1:185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
70.  Rüthrich MM, Giesen N, Mellinghoff SC, Rieger CT, von Lilienfeld-Toal M. Cellular Immune Response after Vaccination in Patients with Cancer-Review on Past and Present Experiences. Vaccines (Basel). 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
71.  Ehmsen S, Asmussen A, Jeppesen SS, Nilsson AC, Østerlev S, Vestergaard H, Justesen US, Johansen IS, Frederiksen H, Ditzel HJ. Antibody and T cell immune responses following mRNA COVID-19 vaccination in patients with cancer. Cancer Cell. 2021;39:1034-1036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 117]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
72.  Mairhofer M, Kausche L, Kaltenbrunner S, Ghanem R, Stegemann M, Klein K, Pammer M, Rauscher I, Salzer HJF, Doppler S, Habringer A, Paar C, Kimeswenger S, Hoetzenecker W, Lamprecht B, Lee S, Schmitt CA. Humoral and cellular immune responses in SARS-CoV-2 mRNA-vaccinated patients with cancer. Cancer Cell. 2021;39:1171-1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
73.  Oosting SF, van der Veldt AAM, GeurtsvanKessel CH, Fehrmann RSN, van Binnendijk RS, Dingemans AC, Smit EF, Hiltermann TJN, den Hartog G, Jalving M, Westphal TT, Bhattacharya A, van der Heiden M, Rimmelzwaan GF, Kvistborg P, Blank CU, Koopmans MPG, Huckriede ALW, van Els CACM, Rots NY, van Baarle D, Haanen JBAG, de Vries EGE. mRNA-1273 COVID-19 vaccination in patients receiving chemotherapy, immunotherapy, or chemoimmunotherapy for solid tumours: a prospective, multicentre, non-inferiority trial. Lancet Oncol. 2021;22:1681-1691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 106]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
74.  Schmidt AL, Labaki C, Hsu CY, Bakouny Z, Balanchivadze N, Berg SA, Blau S, Daher A, El Zarif T, Friese CR, Griffiths EA, Hawley JE, Hayes-Lattin B, Karivedu V, Latif T, Mavromatis BH, McKay RR, Nagaraj G, Nguyen RH, Panagiotou OA, Portuguese AJ, Puc M, Santos Dutra M, Schroeder BA, Thakkar A, Wulff-Burchfield EM, Mishra S, Farmakiotis D, Shyr Y, Warner JL, Choueiri TK; COVID-19 and Cancer Consortium. COVID-19 vaccination and breakthrough infections in patients with cancer. Ann Oncol. 2022;33:340-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 64]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
75.  Goshen-Lago T, Waldhorn I, Holland R, Szwarcwort-Cohen M, Reiner-Benaim A, Shachor-Meyouhas Y, Hussein K, Fahoum L, Baruch M, Peer A, Reiter Y, Almog R, Halberthal M, Ben-Aharon I. Serologic Status and Toxic Effects of the SARS-CoV-2 BNT162b2 Vaccine in Patients Undergoing Treatment for Cancer. JAMA Oncol. 2021;7:1507-1513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 114]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
76.  Waldhorn I, Holland R, Goshen-Lago T, Shirman Y, Szwarcwort-Cohen M, Reiner-Benaim A, Shachor-Meyouhas Y, Hussein K, Fahoum L, Peer A, Almog R, Shaked Y, Halberthal M, Ben-Aharon I. Six-Month Efficacy and Toxicity Profile of BNT162b2 Vaccine in Cancer Patients with Solid Tumors. Cancer Discov. 2021;11:2430-2435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
77.  Barrière J, Chamorey E, Adjtoutah Z, Castelnau O, Mahamat A, Marco S, Petit E, Leysalle A, Raimondi V, Carles M. Impaired immunogenicity of BNT162b2 anti-SARS-CoV-2 vaccine in patients treated for solid tumors. Ann Oncol. 2021;32:1053-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 101]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
78.  Shroff RT, Chalasani P, Wei R, Pennington D, Quirk G, Schoenle MV, Peyton KL, Uhrlaub JL, Ripperger TJ, Jergović M, Dalgai S, Wolf A, Whitmer R, Hammad H, Carrier A, Scott AJ, Nikolich-Žugich J, Worobey M, Sprissler R, Dake M, LaFleur BJ, Bhattacharya D. Immune responses to two and three doses of the BNT162b2 mRNA vaccine in adults with solid tumors. Nat Med. 2021;27:2002-2011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 142]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
79.  Wagner A, Garner-Spitzer E, Schötta AM, Orola M, Wessely A, Zwazl I, Ohradanova-Repic A, Weseslindtner L, Tajti G, Gebetsberger L, Kratzer B, Tomosel E, Kutschera M, Tobudic S, Pickl WF, Kundi M, Stockinger H, Novacek G, Reinisch W, Zielinski C, Wiedermann U. SARS-CoV-2-mRNA Booster Vaccination Reverses Non-Responsiveness and Early Antibody Waning in Immunocompromised Patients-A Phase Four Study Comparing Immune Responses in Patients With Solid Cancers, Multiple Myeloma and Inflammatory Bowel Disease. Front Immunol. 2022;13:889138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 25]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
80.  Reimann P, Ulmer H, Mutschlechner B, Benda M, Severgnini L, Volgger A, Lang T, Atzl M, Huynh M, Gasser K, Grabher C, Mink S, Fraunberger P, Petrausch U, Hartmann B, Winder T. Efficacy and safety of heterologous booster vaccination with Ad26.COV2.S after BNT162b2 mRNA COVID-19 vaccine in haemato-oncological patients with no antibody response. Br J Haematol. 2022;196:577-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
81.  Thomas SJ, Perez JL, Lockhart SP, Hariharan S, Kitchin N, Bailey R, Liau K, Lagkadinou E, Türeci Ö, Şahin U, Xu X, Koury K, Dychter SS, Lu C, Gentile TC, Gruber WC. Efficacy and safety of the BNT162b2 mRNA COVID-19 vaccine in participants with a history of cancer: subgroup analysis of a global phase 3 randomized clinical trial. Vaccine. 2022;40:1483-1492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
82.  Lee LYW, Starkey T, Ionescu MC, Little M, Tilby M, Tripathy AR, Mckenzie HS, Al-Hajji Y, Barnard M, Benny L, Burnett A, Cattell EL, Charman J, Clark JJ, Khan S, Ghafoor Q, Illsley G, Harper-Wynne C, Hattersley RJ, Lee AJX, Leonard PC, Liu JKH; NCRI Consumer Forum, Pang M, Pascoe JS, Platt JR, Potter VA, Randle A, Rigg AS, Robinson TM, Roques TW, Roux RL, Rozmanowski S, Tuthill MH, Watts I, Williams S, Iveson T, Lee SM, Middleton G, Middleton M, Protheroe A, Fittall MW, Fowler T, Johnson P. Vaccine effectiveness against COVID-19 breakthrough infections in patients with cancer (UKCCEP): a population-based test-negative case-control study. Lancet Oncol. 2022;23:748-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 48]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
83.  Thakkar A, Pradhan K, Duva B, Carreno JM, Sahu S, Thiruthuvanathan V, Campbell S, Gallego S, Bhagat TD, Rivera J, Choudhary G, Olea R, Sabalza M, Shapiro LC, Lee M, Quinn R, Mantzaris I, Chu E, Will B, Pirofski LA, Krammer F, Verma A, Halmos B. Study of efficacy and longevity of immune response to third and fourth doses of COVID-19 vaccines in patients with cancer: A single arm clinical trial. Elife. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
84.  Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Perez JL, Pérez Marc G, Moreira ED, Zerbini C, Bailey R, Swanson KA, Roychoudhury S, Koury K, Li P, Kalina WV, Cooper D, Frenck RW Jr, Hammitt LL, Türeci Ö, Nell H, Schaefer A, Ünal S, Tresnan DB, Mather S, Dormitzer PR, Şahin U, Jansen KU, Gruber WC; C4591001 Clinical Trial Group. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med. 2020;383:2603-2615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10556]  [Cited by in F6Publishing: 9515]  [Article Influence: 2378.8]  [Reference Citation Analysis (1)]
85.  Haynes L. Aging of the Immune System: Research Challenges to Enhance the Health Span of Older Adults. Front Aging. 2020;1:602108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
86.  Zangemeister-Wittke U, Simon HU.   Myelosuppression. In: Schwab M. Encyclopedia of Cancer. Berlin, Heidelberg: Springer, 2011: 2437-2440.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Ba Y, Shi Y, Jiang W, Feng J, Cheng Y, Xiao L, Zhang Q, Qiu W, Xu B, Xu R, Shen B, Luo Z, Xie X, Chang J, Wang M, Li Y, Shuang Y, Niu Z, Liu B, Zhang J, Zhang L, Yao H, Xie C, Huang H, Liao W, Chen G, Zhang X, An H, Deng Y, Gong P, Xiong J, Yao Q, An X, Chen C, Wang J, Wang X, Wang Z, Xing P, Yang S, Zhou C. Current management of chemotherapy-induced neutropenia in adults: key points and new challenges: Committee of Neoplastic Supportive-Care (CONS), China Anti-Cancer Association Committee of Clinical Chemotherapy, China Anti-Cancer Association. Cancer Biol Med. 2020;17:896-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
88.  Weinblatt ME, Coblyn JS, Fox DA, Fraser PA, Holdsworth DE, Glass DN, Trentham DE. Efficacy of low-dose methotrexate in rheumatoid arthritis. N Engl J Med. 1985;312:818-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 632]  [Cited by in F6Publishing: 617]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
89.  Alqarni AM, Zeidler MP. How does methotrexate work? Biochem Soc Trans. 2020;48:559-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
90.  Koźmiński P, Halik PK, Chesori R, Gniazdowska E. Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 135]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
91.  Kapetanovic MC, Roseman C, Jönsson G, Truedsson L, Saxne T, Geborek P. Antibody response is reduced following vaccination with 7-valent conjugate pneumococcal vaccine in adult methotrexate-treated patients with established arthritis, but not those treated with tumor necrosis factor inhibitors. Arthritis Rheum. 2011;63:3723-3732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 111]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
92.  Kapetanovic MC, Saxne T, Sjöholm A, Truedsson L, Jönsson G, Geborek P. Influence of methotrexate, TNF blockers and prednisolone on antibody responses to pneumococcal polysaccharide vaccine in patients with rheumatoid arthritis. Rheumatology (Oxford). 2006;45:106-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 233]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
93.  Nived P, Pettersson Å, Jönsson G, Bengtsson AA, Settergren B, Skattum L, Johansson Å, Kapetanovic MC. Methotrexate reduces circulating Th17 cells and impairs plasmablast and memory B cell expansions following pneumococcal conjugate immunization in RA patients. Sci Rep. 2021;11:9199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
94.  Ogino MH, Tadi P.   Cyclophosphamide. 2023 Jul 3. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Gauthier SA, Weiner HL. Cyclophosphamide therapy for MS. Int MS J. 2005;12:52-58.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Elkhalifa A, Weiner H. Cyclophosphamide Treatment of MS: Current Therapeutic Approaches and Treatment Regimens. Int MS J. 2010;17:12-18.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Ahlmann M, Hempel G. The effect of cyclophosphamide on the immune system: implications for clinical cancer therapy. Cancer Chemother Pharmacol. 2016;78:661-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 254]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
98.  Madondo MT, Quinn M, Plebanski M. Low dose cyclophosphamide: Mechanisms of T cell modulation. Cancer Treat Rev. 2016;42:3-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 108]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
99.  de Lavallade H, Khoder A, Hart M, Sarvaria A, Sekine T, Alsuliman A, Mielke S, Bazeos A, Stringaris K, Ali S, Milojkovic D, Foroni L, Chaidos A, Cooper N, Gabriel I, Apperley J, Belsey S, Flanagan RJ, Goldman J, Shpall EJ, Kelleher P, Marin D, Rezvani K. Tyrosine kinase inhibitors impair B-cell immune responses in CML through off-target inhibition of kinases important for cell signaling. Blood. 2013;122:227-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]