ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

COVID-19: Considerations in patients with cancer

COVID-19: Considerations in patients with cancer

INTRODUCTION — At the end of 2019, a novel coronavirus was identified as the cause of a cluster of pneumonia cases in Wuhan, a city in the Hubei Province of China. It rapidly spread, resulting in an epidemic throughout China, followed by an increasing number of cases in other countries throughout the world. In February 2020, the World Health Organization designated the disease COVID-19, which stands for coronavirus disease 2019 [1]. The virus that causes COVID-19 is designated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).

The rapidly expanding COVID-19 pandemic impacted all areas of daily life, including medical care [2]. In particular, delivering care for patients with cancer or suspected cancer during the crisis was challenging given the competing risks of death from untreated cancer versus serious complications from SARS-CoV-2, and the likely higher lethality of COVID-19 in immunocompromised hosts [3-5]. With the generation of effective vaccines and treatments for individuals with COVID-19, many aspects of medical treatment have returned to previous standards. Nevertheless, certain principles related to management of the patient with cancer and COVID-19 remain clinically relevant and will be reviewed here.

Other topics discuss more general issues related to diagnosis and management of COVID-19, and vaccination against SARS-CoV-2.

(See "COVID-19: Diagnosis".)

(See "COVID-19: Evaluation of adults with acute illness in the outpatient setting" and "COVID-19: Management of adults with acute illness in the outpatient setting".)

(See "COVID-19: Management in hospitalized adults".)

(See "COVID-19: Vaccines".)

IMPACT OF THE PANDEMIC ON CANCER SCREENING — Delays in cancer screening during the pandemic led to delayed diagnoses, a higher rate of patients diagnosed in an emergency setting, more diagnoses of later-stage cancers with higher tumor burden, and delays in effective treatment for patients with newly diagnosed malignancies [6-16].

Observational data suggest that cancer-specific mortality was higher during the COVID-19 pandemic when compared with pre-pandemic levels. In the United States, the number of cancer-related deaths increased by 3 percent between 2019 (prior to the pandemic) and 2020 (during the pandemic), with higher cancer mortality rates observed during the times when health care capacity was most challenged by the pandemic [17]. Delays in cancer screening and surveillance during the pandemic may also continue to impact future cancer mortality [18,19].

At present, screening programs have resumed in most areas, with clinicians maintaining full adherence to guidelines for limiting the spread of SARS-CoV-2 infection.

CANCER TREATMENT IN UNINFECTED PATIENTS — The approach to cancer treatment in patients uninfected by SARS-CoV-2 has at this point largely returned to pre-pandemic standards. However, for individuals with a known SARS-CoV-2 exposure, particularly those who are not up to date on COVID-19 vaccination or are expected to have an inadequate immune response to vaccination, it is generally recommended to hold treatment until it is clear that the patient will not develop COVID-19 from that exposure. An exception to holding therapy may be made for patients receiving low-risk cancer therapies, such as hormonal treatments.

Precautions to prevent transmission during routine cancer care are appropriate. (See "COVID-19: General approach to infection prevention in the health care setting".)

Given the risk of transmission from patients with asymptomatic infection, some institutions in areas of high viral transmission are routinely testing all cancer patients prior to all immunosuppressive therapies [20-23], in accordance with guidelines from the Infectious Disease Society of America [24]. According to these guidelines, immunosuppressive procedures are defined as cytotoxic chemotherapy, solid organ or hematopoietic cell transplantation, long-acting biologic therapy, cellular immunotherapy, or high-dose corticosteroids. However, this is not a widespread practice, and it is not supported by guidelines from the American Cancer Society, American Society of Clinical Oncology, or the European Society for Medical Oncology [25,26].

Testing in patients who have undergone laryngectomy is discussed below. (See 'Laryngectomized individuals' below.)

APPROACH TO CANCER PATIENTS WITH COVID-19

COVID-19 outcomes among cancer patients and survivors — Most studies suggest a higher risk of severe COVID-19 in adult patients with active cancer [27-30], although data are mixed [31-33] and outcomes have improved with better COVID-19 therapy and earlier diagnosis [34]. Moreover, many studies were performed prior to availability of effective COVID-19 vaccines.

The risk likely varies by type and stage of cancer and treatment received. In particular, the following features have been associated with an increased risk:

Hematologic malignancies or lung cancer [35-46].

Advanced and/or progressive cancer [28,35,47-53].

Active chemotherapy treatment, particularly more myelosuppressive regimens, has also been described [44,46,54-56], although data are mixed [51,52,57-59]. By contrast, immunotherapy does not appear to worsen outcomes from COVID-19 [52,54,58,60-65], although data in this setting are also conflicting [66,67].

Older age [27,30-32,36,43,45,51,52,58,66] and comorbid conditions [27,30,32,35,36,47-51,57,58,66,68-87] that are independently associated with severe COVID-19 further contribute to the risk in patients with cancer.

However, at least one large meta-analysis has suggested that younger patients with cancer are at higher risk of worsened outcomes from COVID-19 relative to age-matched controls without cancer [44].

Some data suggest that being a survivor of a prior cancer is also a risk factor for severe COVID-19, but the risk is lower compared with active cancer [35,75,88], while other studies have not found higher risks among survivors [51,89].

As an example of available data, in an analysis from the United Kingdom, including records of over 17 million individuals linked to over 10,000 deaths from COVID-19, multivariate analysis found that patients with nonhematologic malignancy diagnosed within one year prior to COVID-19 had a 1.8-fold higher risk of death relative to patients without cancer, and a hematologic malignancy carried a fourfold higher risk [35]. Relative to those diagnosed with cancer within the preceding year, the risks were lower for patients diagnosed with cancer 1 to 4.9 years prior to COVID-19, but still elevated compared with people without cancer; beyond five years, risks for death remained elevated for those with hematologic but not nonhematologic malignancies.

Management of COVID-19 — Overall, the management of COVID-19 in cancer patients is similar to the management used for the general population. However, cancer is considered to be a risk factor for progression to severe COVID-19 (table 1), which influences available treatment options.

Cancer patients in the outpatient setting – For cancer patients in the outpatient setting with COVID-19, available treatment options (algorithm 1) include antiviral agents such as nirmatrelvir-ritonavir and remdesivir. Previously, monoclonal antibodies were also an option; however, they are not active against increasingly prevalent new Omicron variants worldwide. Further details are discussed separately. (See "COVID-19: Management of adults with acute illness in the outpatient setting", section on 'Treatment with COVID-19-specific therapies'.)

Hospitalized cancer patients – The management of hospitalized cancer patients with more severe COVID-19 is the same as that used for the general population and is discussed separately (algorithm 2). (See "COVID-19: Management in hospitalized adults", section on 'COVID-19-specific therapy'.)

If available, convalescent plasma is an additional treatment strategy for hospitalized patients with hematologic malignancies and severe COVID-19 infection. Administration of convalescent plasma is discussed in detail separately. (See "COVID-19: Convalescent plasma and hyperimmune globulin".)

In a randomized, open-label trial in 134 hospitalized patients with severe COVID-19, including 56 patients with cancer, the addition of convalescent/vaccinated plasma to standard of care therapy for COVID-19 did not improve outcomes in the overall population. However, among patients with cancer, convalescent plasma in addition to standard of care resulted in a faster median time to improvement (13 versus 31 days, hazard ratio [HR] 2.50, 95% CI 1.3-4.8) and improved survival (89 versus 57 percent survival at 60 days, HR 0.28, 95% CI 0.06-0.96), relative to standard of care alone [90]. Notably, most patients in this study with cancer had hematologic malignancies (eg, chronic lymphocytic leukemia, acute myelogenous leukemia/myelodysplastic syndrome, non-Hodgkin lymphoma, multiple myeloma), while only a minority (16 percent) had solid tumors. It is possible that some of the benefit observed in patients with hematologic malignancies may be related to their baseline hypogammaglobulinemia (either from their cancer or cancer therapy) and receipt of polyclonal immunoglobulins within the convalescent plasma.

Thrombosis risk – Patients with cancer do not need to take specific precautions against thromboembolism if they develop COVID-19. Although both COVID-19 and cancer predispose towards hypercoagulability, available evidence suggests that cancer patients who develop COVID-19 are not at a higher risk of clotting from COVID-19 than those without cancer. As an example, in a small study in patients with COVID-19, patients with active cancer had a similar risk of thrombotic events as those without cancer (either arterial or venous) at 28 days (14 versus 18 percent) [91]. Issues surrounding anticoagulation for COVID-19 hypercoagulable state are addressed in detail separately. (See "COVID-19: Hypercoagulability", section on 'Management'.)

Persistent infection in immunocompromised patients – Active, persistent SARS-CoV-2 infection can uncommonly occur in immunocompromised patients, particularly those with severe B-cell depletion due to cancer therapy (eg, rituximab, hematopoietic cell transplantation). Such patients typically test positive for SARS CoV-2 on reverse-transcriptase polymerase chain reaction for prolonged periods of time (weeks to months) with a low cycle threshold (which suggests a high viral RNA level). These findings may indicate replicating, transmissible virus. Genomic studies have distinguished such persistent infection from reinfection with prevalent variants by the pattern of viral evolution [92]. (See "COVID-19: Diagnosis", section on 'Cycle threshold' and "COVID-19: Diagnosis", section on 'Diagnosis of reinfection'.)

Data are limited for the optimal management of these patients, and clinical practice is variable. Persistent infection can often be difficult to treat due to the accelerated pace of viral evolution and development of escape mutations [93-96]. Observational studies suggest some efficacy with the combination of antiviral therapy (eg, remdesivir) and passive immunization (eg, convalescent plasma) [93,97-99]. The efficacy of these and other COVID-19 treatments is discussed separately. (See "COVID-19: Management of adults with acute illness in the outpatient setting", section on 'Treatment with COVID-19-specific therapies'.)

Cancer therapy in infected patients

General principles — In the event of a positive SARS-CoV-2 test result, decisions regarding anticancer therapy should be individualized. General principles are as follows:

Systemic therapy

For most patients, chemotherapy or immunotherapy should be interrupted, whether patients are symptomatic from COVID-19 or not [100,101]. However, in cases where expeditious cancer treatment is necessary and patients are asymptomatic or have only mild symptoms of COVID-19, some cancer centers have infusion rooms dedicated to patients who have tested positive for COVID-19 or have had an exposure [102]. Limited data also suggest worsened COVID-19 outcomes with administration of human granulocyte colony-stimulating factor [103].

Nonimmunosuppressive therapies such as hormonal therapies (eg, for breast and prostate cancer) and some oral targeted therapies typically may be continued [104]. Case reports have suggested safety with continuation of anaplastic lymphoma kinase- and c-ROS oncogene 1-targeted therapies among those with the relevant cancer genotypes and COVID-19 pneumonia [105].

Although data are limited, observational studies in chronic lymphocytic leukemia have suggested that Bruton tyrosine kinase inhibitors may be associated with less severe infection [106-109], and continuation of this class of drugs should be considered on a case-by-case basis [110].

Decisions regarding systemic glucocorticoids must be individualized, depending on the dose and indication for the glucocorticoid. As an example, for those with an immunotherapy-related adverse event, it may be reasonable to continue glucocorticoid treatment; by contrast, in a patient with nausea, glucocorticoids could be omitted or an alternative therapy provided. The role of glucocorticoids in COVID-19 management is discussed separately. (See "COVID-19: Management in hospitalized adults", section on 'Dexamethasone and other glucocorticoids' and "COVID-19: Management of adults with acute illness in the outpatient setting", section on 'Therapies of limited or uncertain benefit'.)

Radiation therapy – Cancellation or delay in radiation may be appropriate for patients with COVID-19, after a reassessment of the patient's goals of care. Other potential options, such as modifications to the radiation therapy schedule (eg, hypofractionation), may be considered.

Surgery – The risks of perioperative morbidity and mortality are increased in patients with COVID-19, and the decision to perform surgery must balance this risk against the risks of delaying or avoiding the planned procedure.

When can cancer treatment be safely restarted? — For patients who have discontinued cancer therapy due to infection with COVID-19, we typically resume cancer-specific treatment once transmission-based precautions can be discontinued. More detailed information on discontinuing precautions in immunocompromised patients are presented separately. (See "COVID-19: Infection prevention for persons with SARS-CoV-2 infection", section on 'Immunocompromised patients with confirmed infection'.)

However, earlier resumption of certain therapies (eg, oral chemotherapy or targeted agents, if they have been held) is reasonable, if COVID-19 symptoms are mild and/or improving.

COVID-19 VACCINATION

Whom to vaccinate

Vaccination for cancer patients and survivors — All individuals with active or prior cancer should receive up to date COVID-19 vaccination to prevent SARS-CoV-2 infection. Cancer patients with moderate to severe immunocompromising conditions may have attenuated immunogenicity to the COVID vaccines, but vaccination should still occur in this population. Examples of such moderate to severe immunocompromising conditions include active chemotherapy, hematologic malignancies, hematopoietic stem cell or solid organ transplant, or the use of immunosuppressive medications (eg, rituximab, prolonged use of glucocorticoids). Specific recommendations for COVID-19 vaccination schedules are discussed separately. (See "COVID-19: Vaccines", section on 'Immunocompromised individuals' and "COVID-19: Vaccines", section on 'Dose and interval (for immunocompetent individuals)'.)

If vaccine supply is limited, guidelines suggest prioritizing patients with active cancer who are either on or planning to start treatment (including hematopoietic cell transplant [HCT] and cellular therapies), and those within six months of treatment, except for those receiving only hormonal therapy. Additional factors linked to adverse outcomes from COVID-19, which may increase the priority for vaccination, include age, comorbidities, and sociodemographic factors (eg, poverty, limited access to health care, and under-represented minorities [45]). (See 'COVID-19 outcomes among cancer patients and survivors' above.)

Assessment for potential vaccine contraindications — This subject is discussed in detail separately. (See "COVID-19: Allergic reactions to SARS-CoV-2 vaccines", section on 'Uncertain role of polyethylene glycol'.)

Thrombotic risks are not a contraindication — Although single-dose adenoviral vaccines have been associated with a rare risk of thrombosis with thrombocytopenia after vaccination, no risk factors for this have been identified. Therefore, a prior history of venous thromboembolism (VTE), or predisposition to VTE, is not a contraindication to vaccination with any type of vaccine. (See "COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT)", section on 'Individuals with thrombotic risk factors, prior thrombosis, or prior HIT'.)

Safety and efficacy — Available data suggest that COVID-19 vaccination is safe in patients with cancer and reduces the risk of SARS-CoV-2 infection and long-term sequelae [111-114], although the risk of breakthrough infections may be higher than in those without cancer.

As examples,

One study found that the vaccine was 58 percent effective at preventing SARS-CoV-2 infections in patients with cancer, two weeks after the second dose [112]. However, studies also suggest that vaccine efficacy is reduced in those with active cancer relative to those without cancer, especially hematologic malignancies and particularly those on anti-CD20 antibody treatment [115-132].

In a registry study including 6860 partially or full vaccinated patients with an mRNA vaccine who developed COVID-19, including 1460 patients with cancer, cancer was associated with higher risks for breakthrough infection (odds ratio [OR] 1.1) and severe outcomes (OR 1.3) compared with not having cancer, after adjustment for other clinical factors [131]. Compared with solid tumors, hematologic malignancies were at increased risk for breakthrough infections (eg, adjusted OR for lymphoma 2.1; and for lymphoid leukemia 7.3). Breakthrough risks were lower for those who received a second dose of the vaccine, as well as for those who received the mRNA-1273 (Moderna COVID-19) rather than the BNT162b2 (Pfizer COVID-19) vaccination.

A study in 45,000 vaccinated patients with cancer suggested that the risk of breakthrough infections in patients with all cancer was approximately 14 percent, with highest risk for pancreatic (25 percent), liver (23 percent), lung (20 percent), and colorectal (18 percent) cancers; and lowest risks for thyroid (10 percent), endometrial (12 percent), and breast (12 percent) cancers versus 4.9 percent in the noncancer population [133].

Immunogenicity studies also suggest decreased immune response among cancer patients, particularly those with hematologic malignancies [121,122,134-138]. As an example, in a study of 200 patients with cancer who had been vaccinated (with either two doses of the mRNA vaccines or one dose of the adenoviral vaccine), the seroconversion rate was 94 percent overall, 98 percent among those with solid tumors, and 85 percent in those with a hematologic malignancy [122]. Among those receiving anti-CD20 therapies or following HCT, the seroconversion rate was approximately 70 percent. Separately, an inverse association has been described between the SARS-CoV-2 spike protein antibody vaccine response and infection severity in patients with cancer [139].

Overall, these data underscore the need to maintain appropriate precautions in all patients with cancer, and to not delay the timing of additional doses, if possible [126,128,140-149].

There are limited studies directly comparing the available vaccines in cancer patients. Although most evaluated the use of mRNA vaccines (BNT162b2 [Pfizer COVID-19 vaccine] and mRNA-1273 [Moderna COVID-19 vaccine]), observational data in patients with solid organ or hematologic cancers suggest that one dose of the Ad26.COV2.S (Janssen COVID-19 vaccine) is associated with lower protective immune responses compared with two doses of the mRNA vaccines [117,120], which has been also seen in the general population. (See "COVID-19: Vaccines", section on 'Approach to vaccination in the United States'.)

None of these vaccines can cause SARS-CoV-2 infection, regardless of immunosuppression. In contrast to other vaccines that consist of live or attenuated virus, none of the currently available COVID-19 vaccines contain infectious SARS-CoV-2. Although Ad26.COV2.S (Janssen COVID-19 vaccine) uses an adenoviral vector platform, the adenovirus is nonreplicative. (See "Immunizations in adults with cancer", section on 'General approach' and "COVID-19: Vaccines", section on 'General principles'.)

Timing

Relative to therapy — For patients receiving immunosuppressive therapy, our approach is to administer the vaccination between treatment cycles, when immunosuppression from treatment is minimized. However, for patients with marrow failure from disease and/or therapy who are expected to have limited or no recovery, as well as those receiving continuous treatment with targeted agents, vaccination should be administered when it is available. This approach is generally consistent with that from expert groups [150,151].

For those who received COVID-19 vaccination prior to HCT or chimeric antigen receptor modified T (CAR-T) cell therapy, the CDC recommends repeat vaccination with a full primary series at least three months after the transplant or CAR-T administration. (See "Immunizations in hematopoietic cell transplant candidates and recipients".)

Some expert groups recommend holding certain immunosuppressive agents around the time of vaccination or adjusting the timing of vaccination to account for receipt of such agents to try to optimize the vaccine response. As an example, for patients receiving rituximab, the American College of Rheumatology suggests scheduling vaccination so that the series is initiated approximately four weeks prior to the next scheduled rituximab dose and delaying administration of rituximab until two to four weeks after completion of vaccination, if disease activity allows [152]. (See "COVID-19: Care of adult patients with systemic rheumatic disease", section on 'Coordinating vaccine administration with immunosuppressive therapy'.)

Relative to radiologic imaging — While previous recommendations suggested scheduling imaging prior to a dose of a COVID-19 mRNA vaccine, or four to six weeks after a dose, we no longer alter imaging schedules around COVID vaccination schedule; rather, radiologists take into consideration COVID vaccination history in interpretation of imaging findings.

Further details regarding mammography for breast cancer screening in the context of COVID vaccination are found elsewhere. (See "Screening for breast cancer: Strategies and recommendations", section on 'Recent COVID-19 vaccination'.)

Vaccine-associated lymphadenopathy — For cancer patients and survivors with an identified primary tumor site (eg, breast cancer, melanoma, head and neck cancer), it is preferable to administer the COVID-19 vaccine into the arm contralateral to the primary tumor to avoid confusing benign vaccine-associated adenopathy with malignant adenopathy on imaging studies [153-155]. By using this approach, lymphadenopathy that is detected after vaccination can often be observed for resolution, potentially reducing the need for further diagnostic imaging and biopsies.

Axillary swelling or tenderness has been reported in approximately 12 percent of patients after the first dose of mRNA-1273 (Moderna COVID-19 vaccine) and in 16 percent after the second dose [156]. Lymphadenopathy of the arm and neck has also been reported as an unsolicited event in 1 percent or fewer patients [157]. Clinically symptomatic lymphadenopathy generally developed within two to four days of vaccination and resolved within two weeks by exam [157], but can persist subclinically for weeks on imaging, with one study reporting a median of 97 days [156].

Reported rates and duration of lymphadenopathy in both trials were based upon clinical assessment, and therefore, the rates of subclinical adenopathy detected by radiologic imaging might be higher, as high as 53 percent in one study, in which most of the imaging was performed by positron emission tomography/computed tomography [158].

Impact of immune checkpoint inhibitors — We do not consider immune checkpoint inhibitors (ICIs), which stimulate immune system function, to be a contraindication to COVID-19 vaccination. Available data on patients with cancer who received COVID-19 vaccinations and are treated with ICIs suggest that vaccination is effective and well tolerated [145,148,159], without increased risk of immune-related adverse events [160]. It is also likely that the benefits of vaccination in preventing severe COVID-19 infection outweigh the potential risks in this population.

Is there a way to assess for successful immunization? — Although some antibody titers may correlate with vaccine efficacy, they cannot be reliably used to assess for immunity against COVID-19 following vaccination. Further details on assessing postvaccination serologies are discussed separately. (See "COVID-19: Vaccines", section on 'Limited role for post-vaccination testing' and "COVID-19: Diagnosis", section on 'Testing following COVID-19 vaccination'.)

SPECIAL CONSIDERATIONS

Differentiating lymphangitic spread, pneumonitis, and COVID-19 — Some systemic cancer treatments are associated with a risk of pneumonitis (eg, immune checkpoint inhibitors, gemcitabine, mechanistic [previously referred to as mammalian] target of rapamycin inhibitors). In other cases, new infiltrates on radiographic imaging may reflect disease progression (eg, lymphangitic spread) or radiation pneumonitis [161]. (See "Toxicities associated with immune checkpoint inhibitors", section on 'Pneumonitis' and "Pulmonary toxicity associated with antineoplastic therapy: Molecularly targeted agents".)

Besides the fact that treatment-related pneumonitis might increase the risk of serious complications if the patient develops COVID-19, it may be difficult to distinguish therapy effect versus disease progression versus viral infection. In this setting, cancer treatment should be held until the diagnosis is clarified. Testing for COVID-19 is appropriate in such circumstances, similar to the approach taken for patients with new respiratory symptoms. (See "COVID-19: Diagnosis".)

If testing for COVID-19 is negative, other diagnostic and therapeutic interventions for potential drug induced pneumonitis may be pursued. (See "Toxicities associated with immune checkpoint inhibitors", section on 'Pneumonitis' and "Pulmonary toxicity associated with systemic antineoplastic therapy: Clinical presentation, diagnosis, and treatment".)

Incorporation of telehealth — The benefits of virtual visits in oncology care include enabling care (including the maintenance of participation in clinical trials) during the pandemic while avoiding communicable disease exposure, increased patient access, and convenience. Accumulating data support mostly favorable outcomes in cancer patients [162-169]. Further discussion of telehealth is found separately. (See "Telemedicine for adults".)

Limited roles for pre- and postexposure prophylaxis — There is no role for pre-exposure prophylaxis with monoclonal antibodies in individuals at high risk for severe COVID-19 (eg, solid organ and hematopoietic cell transplant patients), even if they are expected to have suboptimal protection from vaccination (table 2), or are unable to receive vaccination. This is due to inactivity against the most common Omicron subvariants. Similarly, there is only a limited role for monoclonal antibodies as post-exposure prophylaxis. Further details are discussed separately. (See "COVID-19: Epidemiology, virology, and prevention", section on 'Monoclonal antibodies ineffective for pre-exposure prophylaxis' and "COVID-19: Epidemiology, virology, and prevention", section on 'No role for post-exposure prophylaxis'.)

Laryngectomized individuals — Some head and neck cancer survivors will have undergone permanent laryngectomy during treatment of their cancer. In general, such patients should wear a highly efficient heat and moisture exchanger over the stoma at all times, especially when around other people. In addition, they can wear a surgical mask (preferably an N95 respirator) over the stoma, and an additional surgical mask or respirator over the nose and mouth [170,171]. (See "Alaryngeal speech rehabilitation".)

When carrying out COVID-19 testing in persons with a laryngectomy, swabbing and analysis from both the stoma and the nose may increase sensitivity [172,173]. At least one case report documents a laryngectomee who had a positive COVID-19 diagnostic test from the nasopharyngeal swab and a negative result from a tracheal swab [174].

SUMMARY AND RECOMMENDATIONS

Cancer screening and surveillance – During the pandemic, delays in cancer screening and diagnostic/surveillance testing were frequent. However, most screening programs have resumed. (See 'Impact of the pandemic on cancer screening' above.)

Cancer treatment in uninfected patients – Cancer care is largely proceeding along the lines of pre-pandemic standards. However, for individuals with a known severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure, particularly those who are not up to date on COVID-19 vaccination or are expected to have an inadequate immune response to vaccination, we typically hold treatment until it is clear that the patient will not develop COVID-19 from that exposure. An exception may be made for patients receiving low-risk therapies, such as hormonal treatments. (See 'Cancer treatment in uninfected patients' above.)

Cancer patients with COVID-19

Risk factors – Most studies suggest a higher risk of severe COVID-19 in adult patients with active cancer, although data are mixed. In particular, hematologic malignancies, lung cancer, advanced or progressive cancer, active chemotherapy, older age, and comorbid conditions are risk factors for severe COVID-19. Prior cancer is also a risk factor, but the risk is lower compared with active cancer. (See 'COVID-19 outcomes among cancer patients and survivors' above.)

Management of COVID-19 – Overall, COVID-19 management for patients with cancer is similar to the management used for the general population. However, cancer is considered to be a risk factor for progression to severe COVID-19 (table 1), which influences available treatment options. (See 'Management of COVID-19' above.)

Cancer therapy in infected patients – For most cancer patients with COVID-19, chemotherapy or immunotherapy should be interrupted, whether patients are symptomatic from COVID-19 or not. We typically resume cancer treatment once transmission-based precautions can be discontinued; the duration of such precautions is often determined by regional and institutional protocol. (See 'Cancer therapy in infected patients' above.)

COVID-19 vaccination

Vaccination in cancer patients – All individuals with active or prior cancer should receive up to date COVID-19 vaccination to prevent SARS-CoV-2 infection. (See "COVID-19: Vaccines", section on 'Immunocompromised individuals' and "COVID-19: Vaccines", section on 'Dose and interval (for immunocompetent individuals)'.)

  1. World Health Organization. Director-General's remarks at the media briefing on 2019-nCoV on 11 February 2020. Available at: http://www.who.int/dg/speeches/detail/who-director-general-s-remarks-at-the-media-briefing-on-2019-ncov-on-11-february-2020 (Accessed on February 12, 2020).
  2. Han X, Shi SK, Zhao J, et al. The first year of the COVID-19 pandemic and health among cancer survivors in the United States. Cancer 2022; 128:3727.
  3. 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.
  4. Lewis MA. Between Scylla and Charybdis - Oncologic Decision Making in the Time of Covid-19. N Engl J Med 2020; 382:2285.
  5. Hui D, Nortje N, George M, et al. Impact of an Interdisciplinary Goals-of-Care Program Among Medical Inpatients at a Comprehensive Cancer Center During the COVID-19 Pandemic: A Propensity Score Analysis. J Clin Oncol 2023; 41:579.
  6. Toss A, Isca C, Venturelli M, et al. Two-month stop in mammographic screening significantly impacts on breast cancer stage at diagnosis and upfront treatment in the COVID era. ESMO Open 2021; 6:100055.
  7. Kuzuu K, Misawa N, Ashikari K, et al. Gastrointestinal Cancer Stage at Diagnosis Before and During the COVID-19 Pandemic in Japan. JAMA Netw Open 2021; 4:e2126334.
  8. Thierry AR, Pastor B, Pisareva E, et al. Association of COVID-19 Lockdown With the Tumor Burden in Patients With Newly Diagnosed Metastatic Colorectal Cancer. JAMA Netw Open 2021; 4:e2124483.
  9. Englum BR, Prasad NK, Lake RE, et al. Impact of the COVID-19 pandemic on diagnosis of new cancers: A national multicenter study of the Veterans Affairs Healthcare System. Cancer 2022; 128:1048.
  10. Sharma J, Mahajan A, Bakhshi S, et al. The impact of COVID-19 pandemic on access to treatment for children with cancer in India and treating center practices. Cancer 2022; 128:579.
  11. AACR Report on the Impact of COVID-19 on Cancer Research and Patient Care. Available at: https://www.aacr.org/wp-content/uploads/2022/02/AACR_C19CR_2022.pdf (Accessed on February 15, 2022).
  12. Eskander A, Li Q, Yu J, et al. Incident Cancer Detection During the COVID-19 Pandemic. J Natl Compr Canc Netw 2022; 20:276.
  13. Joung RH, Nelson H, Mullett TW, et al. A national quality improvement study identifying and addressing cancer screening deficits due to the COVID-19 pandemic. Cancer 2022; 128:2119.
  14. Zhang X, Elsaid MI, DeGraffinreid C, et al. Impact of the COVID-19 Pandemic on Cancer Screening Delays. J Clin Oncol 2023; 41:3194.
  15. Star J, Bandi P, Siegel RL, et al. Cancer Screening in the United States During the Second Year of the COVID-19 Pandemic. J Clin Oncol 2023; 41:4352.
  16. Negoita S, Chen HS, Sanchez PV, et al. Annual Report to the Nation on the Status of Cancer, part 2: Early assessment of the COVID-19 pandemic's impact on cancer diagnosis. Cancer 2024; 130:117.
  17. Zhao J, Han X, Miller KD, et al. Association of the COVID-19 Pandemic and Changes in Patterns of Cancer-Related Mortality in the United States. JCO Oncol Pract 2023; 19:501.
  18. Alagoz O, Lowry KP, Kurian AW, et al. Impact of the COVID-19 Pandemic on Breast Cancer Mortality in the US: Estimates From Collaborative Simulation Modeling. J Natl Cancer Inst 2021; 113:1484.
  19. Teglia F, Angelini M, Astolfi L, et al. Global Association of COVID-19 Pandemic Measures With Cancer Screening: A Systematic Review and Meta-analysis. JAMA Oncol 2022; 8:1287.
  20. Madariaga A, McMullen M, Sheikh S, et al. COVID-19 Testing in Patients with Cancer: Does One Size Fit All? Clin Cancer Res 2020; 26:4737.
  21. Ontario Ministry of Health. COVID-19 Provincial Testing Guidance Update; 2020. Available at: http://www.health.gov.on.ca/en/pro/programs/publichealth/coronavirus/docs/2019_testing_guidance.pdf (Accessed on August 27, 2020).
  22. Martin M, Guerrero-Zotano A, Montero Á, et al. GEICAM Guidelines for the Management of Patients with Breast Cancer During the COVID-19 Pandemic in Spain. Oncologist 2020; 25:e1339.
  23. Al-Shamsi HO, Coomes EA, Alrawi S. Screening for COVID-19 in Asymptomatic Patients With Cancer in a Hospital in the United Arab Emirates. JAMA Oncol 2020; 6:1627.
  24. Infectious Diseases Society of America Guidelines on the Diagnosis of COVID-19. Available at: https://www.idsociety.org/practice-guideline/covid-19-guideline-diagnostics/ (Accessed on May 08, 2020).
  25. Ürün Y, Hussain SA, Bakouny Z, et al. Survey of the Impact of COVID-19 on Oncologists' Decision Making in Cancer. JCO Glob Oncol 2020; 6:1248.
  26. Schultz L, Link MP, Rheingold S, et al. Summary of COVID-19 clinical practice adjustments across select institutions. Pediatr Blood Cancer 2020; 67:e28411.
  27. Lunski MJ, Burton J, Tawagi K, et al. Multivariate mortality analyses in COVID-19: Comparing patients with cancer and patients without cancer in Louisiana. Cancer 2021; 127:266.
  28. Bertuzzi AF, Ciccarelli M, Marrari A, et al. Impact of active cancer on COVID-19 survival: a matched-analysis on 557 consecutive patients at an Academic Hospital in Lombardy, Italy. Br J Cancer 2021; 125:358.
  29. Fu C, Stoeckle JH, Masri L, et al. COVID-19 outcomes in hospitalized patients with active cancer: Experiences from a major New York City health care system. Cancer 2021; 127:3466.
  30. Pinato DJ, Tabernero J, Bower M, et al. Prevalence and impact of COVID-19 sequelae on treatment and survival of patients with cancer who recovered from SARS-CoV-2 infection: evidence from the OnCovid retrospective, multicentre registry study. Lancet Oncol 2021; 22:1669.
  31. Docherty AB, Harrison EM, Green CA, et al. Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study. BMJ 2020; 369:m1985.
  32. Brar G, Pinheiro LC, Shusterman M, et al. COVID-19 Severity and Outcomes in Patients With Cancer: A Matched Cohort Study. J Clin Oncol 2020; 38:3914.
  33. Sengar M, Chinnaswamy G, Ranganathan P, et al. Outcomes of COVID-19 and risk factors in patients with cancer. Nat Cancer 2022; 3:547.
  34. OnCovid Study Group, Pinato DJ, Patel M, et al. Time-Dependent COVID-19 Mortality in Patients With Cancer: An Updated Analysis of the OnCovid Registry. JAMA Oncol 2022; 8:114.
  35. Williamson EJ, Walker AJ, Bhaskaran K, et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature 2020; 584:430.
  36. Mehta V, Goel S, Kabarriti R, et al. Case Fatality Rate of Cancer Patients with COVID-19 in a New York Hospital System. Cancer Discov 2020; 10:935.
  37. Cai Y, Hao Z, Gao Y, et al. Coronavirus Disease 2019 in the Perioperative Period of Lung Resection: A Brief Report From a Single Thoracic Surgery Department in Wuhan, People's Republic of China. J Thorac Oncol 2020; 15:1065.
  38. Luo J, Rizvi H, Preeshagul IR, et al. COVID-19 in patients with lung cancer. Ann Oncol 2020; 31:1386.
  39. Horn L, Whisenant JG, Torri V, et al. Thoracic Cancers International COVID-19 Collaboration (TERAVOLT): Impact of type of cancer therapy and COVID therapy on survival. J Clin Oncol 2020; 38S:ASCO #LBA111.
  40. Johnston EE, Martinez I, Davis ES, et al. SARS-CoV-2 in Childhood Cancer in 2020: A Disease of Disparities. J Clin Oncol 2021; 39:3778.
  41. Whisenant JG, Baena J, Cortellini A, et al. A Definitive Prognostication System for Patients With Thoracic Malignancies Diagnosed With Coronavirus Disease 2019: An Update From the TERAVOLT Registry. J Thorac Oncol 2022; 17:661.
  42. Várnai C, Palles C, Arnold R, et al. Mortality Among Adults With Cancer Undergoing Chemotherapy or Immunotherapy and Infected With COVID-19. JAMA Netw Open 2022; 5:e220130.
  43. Mileham KF, Bruinooge SS, Aggarwal C, et al. Changes Over Time in COVID-19 Severity and Mortality in Patients Undergoing Cancer Treatment in the United States: Initial Report From the ASCO Registry. JCO Oncol Pract 2022; 18:e426.
  44. Khoury E, Nevitt S, Madsen WR, et al. Differences in Outcomes and Factors Associated With Mortality Among Patients With SARS-CoV-2 Infection and Cancer Compared With Those Without Cancer: A Systematic Review and Meta-analysis. JAMA Netw Open 2022; 5:e2210880.
  45. Han X, Hu X, Zhao J, et al. Identification of Deaths Caused by Cancer and COVID-19 in the US During March to December 2020. JAMA Oncol 2022; 8:1696.
  46. Gong IY, Vijenthira A, Powis M, et al. Association of COVID-19 Vaccination With Breakthrough Infections and Complications in Patients With Cancer. JAMA Oncol 2023; 9:386.
  47. Giannakoulis VG, Papoutsi E, Siempos II. Effect of Cancer on Clinical Outcomes of Patients With COVID-19: A Meta-Analysis of Patient Data. JCO Glob Oncol 2020; 6:799.
  48. Dai M, Liu D, Liu M, et al. Patients with Cancer Appear More Vulnerable to SARS-CoV-2: A Multicenter Study during the COVID-19 Outbreak. Cancer Discov 2020; 10:783.
  49. Tian J, Yuan X, Xiao J, et al. Clinical characteristics and risk factors associated with COVID-19 disease severity in patients with cancer in Wuhan, China: a multicentre, retrospective, cohort study. Lancet Oncol 2020; 21:893.
  50. Passamonti F, Cattaneo C, Arcaini L, et al. Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: a retrospective, multicentre, cohort study. Lancet Haematol 2020; 7:e737.
  51. Kuderer NM, Choueiri TK, Shah DP, et al. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet 2020; 395:1907.
  52. Albiges L, Foulon S, Bayle A, et al. Determinants of the outcomes of patients with cancer infected with SARS-CoV-2: results from the Gustave Roussy cohort. Nat Cancer 2020; 1:965.
  53. Pinato DJ, Zambelli A, Aguilar-Company J, et al. Clinical portrait of the SARS-CoV-2 epidemic in European cancer patients. Cancer Discov 2020.
  54. Sharafeldin N, Bates B, Song Q, et al. Outcomes of COVID-19 in Patients With Cancer: Report From the National COVID Cohort Collaborative (N3C). J Clin Oncol 2021; 39:2232.
  55. Clift AK, Coupland CAC, Keogh RH, et al. Living risk prediction algorithm (QCOVID) for risk of hospital admission and mortality from coronavirus 19 in adults: national derivation and validation cohort study. BMJ 2020; 371:m3731.
  56. Chavez-MacGregor M, Lei X, Zhao H, et al. Evaluation of COVID-19 Mortality and Adverse Outcomes in US Patients With or Without Cancer. JAMA Oncol 2022; 8:69.
  57. Jee J, Foote MB, Lumish M, et al. Chemotherapy and COVID-19 Outcomes in Patients With Cancer. J Clin Oncol 2020; 38:3538.
  58. Lee LY, Cazier JB, Angelis V, et al. COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: a prospective cohort study. Lancet 2020; 395:1919.
  59. Fox TA, Troy-Barnes E, Kirkwood AA, et al. Clinical outcomes and risk factors for severe COVID-19 in patients with haematological disorders receiving chemo- or immunotherapy. Br J Haematol 2020; 191:194.
  60. Luo J, Rizvi H, Egger JV, et al. Impact of PD-1 Blockade on Severity of COVID-19 in Patients with Lung Cancers. Cancer Discov 2020; 10:1121.
  61. Fillmore NR, La J, Szalat RE, et al. Prevalence and Outcome of COVID-19 Infection in Cancer Patients: A National Veterans Affairs Study. J Natl Cancer Inst 2021; 113:691.
  62. Garassino MC, Whisenant JG, Huang LC, et al. COVID-19 in patients with thoracic malignancies (TERAVOLT): first results of an international, registry-based, cohort study. Lancet Oncol 2020; 21:914.
  63. Rogiers A, Pires da Silva I, Tentori C, et al. Clinical impact of COVID-19 on patients with cancer treated with immune checkpoint inhibition. J Immunother Cancer 2021; 9.
  64. Klebanov N, Pahalyants V, Murphy WS, et al. Risk of COVID-19 in Patients with Cancer Receiving Immune Checkpoint Inhibitors. Oncologist 2021; 26:e898.
  65. Yekedüz E, Utkan G, Ürün Y. A systematic review and meta-analysis: the effect of active cancer treatment on severity of COVID-19. Eur J Cancer 2020; 141:92.
  66. Robilotti EV, Babady NE, Mead PA, et al. Determinants of COVID-19 disease severity in patients with cancer. Nat Med 2020; 26:1218.
  67. Bakouny Z, Labaki C, Grover P, et al. Interplay of Immunosuppression and Immunotherapy Among Patients With Cancer and COVID-19. JAMA Oncol 2023; 9:128.
  68. Yarza R, Bover M, Paredes D, et al. SARS-CoV-2 infection in cancer patients undergoing active treatment: analysis of clinical features and predictive factors for severe respiratory failure and death. Eur J Cancer 2020; 135:242.
  69. Wang Q, Berger NA, Xu R. Analyses of Risk, Racial Disparity, and Outcomes Among US Patients With Cancer and COVID-19 Infection. JAMA Oncol 2021; 7:220.
  70. Venkatesulu BP, Chandrasekar VT, Girdhar P, et al. A systematic review and meta-analysis of cancer patients affected by a novel coronavirus. medRxiv 2020.
  71. Saini KS, Tagliamento M, Lambertini M, et al. Mortality in patients with cancer and coronavirus disease 2019: A systematic review and pooled analysis of 52 studies. Eur J Cancer 2020; 139:43.
  72. Miyashita H, Mikami T, Chopra N, et al. Do patients with cancer have a poorer prognosis of COVID-19? An experience in New York City. Ann Oncol 2020; 31:1088.
  73. Cook G, John Ashcroft A, Pratt G, et al. Real-world assessment of the clinical impact of symptomatic infection with severe acute respiratory syndrome coronavirus (COVID-19 disease) in patients with multiple myeloma receiving systemic anti-cancer therapy. Br J Haematol 2020; 190:e83.
  74. Singh AK, Gillies CL, Singh R, et al. Prevalence of co-morbidities and their association with mortality in patients with COVID-19: A systematic review and meta-analysis. Diabetes Obes Metab 2020; 22:1915.
  75. van Doesum J, Chinea A, Pagliaro M, et al. Clinical characteristics and outcome of SARS-CoV-2-infected patients with haematological diseases: a retrospective case study in four hospitals in Italy, Spain and the Netherlands. Leukemia 2020; 34:2536.
  76. Ofori-Asenso R, Ogundipe O, Agyeman AA, et al. Cancer is associated with severe disease in COVID-19 patients: a systematic review and meta-analysis. Ecancermedicalscience 2020; 14:1047.
  77. Grasselli G, Greco M, Zanella A, et al. Risk Factors Associated With Mortality Among Patients With COVID-19 in Intensive Care Units in Lombardy, Italy. JAMA Intern Med 2020; 180:1345.
  78. Hultcrantz M, Richter J, Rosenbaum C, et al. COVID-19 infections and outcomes in patients with multiple myeloma in New York City: a cohort study from five academic centers. medRxiv 2020.
  79. Lee LYW, Cazier JB, Starkey T, et al. COVID-19 prevalence and mortality in patients with cancer and the effect of primary tumour subtype and patient demographics: a prospective cohort study. Lancet Oncol 2020; 21:1309.
  80. Mato AR, Roeker LE, Lamanna N, et al. Outcomes of COVID-19 in patients with CLL: a multicenter international experience. Blood 2020; 136:1134.
  81. Zhou Y, Yang Q, Chi J, et al. Comorbidities and the risk of severe or fatal outcomes associated with coronavirus disease 2019: A systematic review and meta-analysis. Int J Infect Dis 2020; 99:47.
  82. Gupta S, Hayek SS, Wang W, et al. Factors Associated With Death in Critically Ill Patients With Coronavirus Disease 2019 in the US. JAMA Intern Med 2020; 180:1436.
  83. Vijenthira A, Gong IY, Fox TA, et al. Outcomes of patients with hematologic malignancies and COVID-19: a systematic review and meta-analysis of 3377 patients. Blood 2020; 136:2881.
  84. Zhang H, Han H, He T, et al. Clinical Characteristics and Outcomes of COVID-19-Infected Cancer Patients: A Systematic Review and Meta-Analysis. J Natl Cancer Inst 2021; 113:371.
  85. Barek MA, Aziz MA, Islam MS. Impact of age, sex, comorbidities and clinical symptoms on the severity of COVID-19 cases: A meta-analysis with 55 studies and 10014 cases. Heliyon 2020; 6:e05684.
  86. Desai A, Gupta R, Advani S, et al. Mortality in hospitalized patients with cancer and coronavirus disease 2019: A systematic review and meta-analysis of cohort studies. Cancer 2021; 127:1459.
  87. Jiang C, Yabroff KR, Deng L, et al. Prevalence of Underlying Medical Conditions Associated With Severe COVID-19 Illness in Adult Cancer Survivors in the United States. J Natl Cancer Inst 2022; 114:156.
  88. Mangone L, Gioia F, Mancuso P, et al. Cumulative COVID-19 incidence, mortality and prognosis in cancer survivors: A population-based study in Reggio Emilia, Northern Italy. Int J Cancer 2021.
  89. Gupta S, Sutradhar R, Alexander S, Science M. Risk of COVID-19 Infections and of Severe Complications Among Survivors of Childhood, Adolescent, and Young Adult Cancer: A Population-Based Study in Ontario, Canada. J Clin Oncol 2022.
  90. Denkinger CM, Janssen M, Schäkel U, et al. Anti-SARS-CoV-2 antibody-containing plasma improves outcome in patients with hematologic or solid cancer and severe COVID-19: a randomized clinical trial. Nat Cancer 2023; 4:96.
  91. Patell R, Bogue T, Bindal P, et al. Incidence of thrombosis and hemorrhage in hospitalized cancer patients with COVID-19. J Thromb Haemost 2020; 18:2349.
  92. Choudhary MC, Crain CR, Qiu X, et al. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Sequence Characteristics of Coronavirus Disease 2019 (COVID-19) Persistence and Reinfection. Clin Infect Dis 2022; 74:237.
  93. Nussenblatt V, Roder AE, Das S, et al. Year-long COVID-19 infection reveals within-host evolution of SARS-CoV-2 in a patient with B cell depletion. medRxiv 2021.
  94. Hensley MK, Bain WG, Jacobs J, et al. Intractable Coronavirus Disease 2019 (COVID-19) and Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Replication in a Chimeric Antigen Receptor-Modified T-Cell Therapy Recipient: A Case Study. Clin Infect Dis 2021; 73:e815.
  95. Scherer EM, Babiker A, Adelman MW, et al. SARS-CoV-2 Evolution and Immune Escape in Immunocompromised Patients. N Engl J Med 2022; 386:2436.
  96. Kemp SA, Collier DA, Datir RP, et al. SARS-CoV-2 evolution during treatment of chronic infection. Nature 2021; 592:277.
  97. Malsy J, Veletzky L, Heide J, et al. Sustained Response After Remdesivir and Convalescent Plasma Therapy in a B-Cell-Depleted Patient With Protracted Coronavirus Disease 2019 (COVID-19). Clin Infect Dis 2021; 73:e4020.
  98. Baang JH, Smith C, Mirabelli C, et al. Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 Replication in an Immunocompromised Patient. J Infect Dis 2021; 223:23.
  99. Dioverti MV, Gaston DC, Morris CP, et al. Combination Therapy With Casirivimab/Imdevimab and Remdesivir for Protracted SARS-CoV-2 Infection in B-cell-Depleted Patients. Open Forum Infect Dis 2022; 9:ofac064.
  100. Curigliano G, Banerjee S, Cervantes A, et al. Managing cancer patients during the COVID-19 pandemic: an ESMO multidisciplinary expert consensus. Ann Oncol 2020; 31:1320.
  101. Sullivan RJ, Johnson DB, Rini BI, et al. COVID-19 and immune checkpoint inhibitors: initial considerations. J Immunother Cancer 2020; 8.
  102. Abedi SM, Lekkala M, Moftakhar B, et al. Protecting High-Risk Oncology Patients During the COVID-19 Pandemic by Creating an Isolated Outpatient Clinic. JCO Oncol Pract 2022; 18:e626.
  103. Zhang AW, Morjaria S, Kaltsas A, et al. The Effect of Neutropenia and Filgrastim (G-CSF) on Cancer Patients With Coronavirus Disease 2019 (COVID-19) Infection. Clin Infect Dis 2022; 74:567.
  104. Schmidt AL, Tucker MD, Bakouny Z, et al. Association Between Androgen Deprivation Therapy and Mortality Among Patients With Prostate Cancer and COVID-19. JAMA Netw Open 2021; 4:e2134330.
  105. Leonetti A, Facchinetti F, Zielli T, et al. COVID-19 in lung cancer patients receiving ALK/ROS1 inhibitors. Eur J Cancer 2020; 132:122.
  106. Reda G, Noto A, Cassin R, et al. Reply to "CLL and COVID-19 at the Hospital Clinic of Barcelona: an interim report" Analysis of six hematological centers in Lombardy : On behalf of CLL commission of Lombardy Hematology Network (REL). Leukemia 2020; 34:2531.
  107. Treon SP, Castillo JJ, Skarbnik AP, et al. The BTK inhibitor ibrutinib may protect against pulmonary injury in COVID-19-infected patients. Blood 2020; 135:1912.
  108. Thibaud S, Tremblay D, Bhalla S, et al. Protective role of Bruton tyrosine kinase inhibitors in patients with chronic lymphocytic leukaemia and COVID-19. Br J Haematol 2020; 190:e73.
  109. Scarfò L, Chatzikonstantinou T, Rigolin GM, et al. COVID-19 severity and mortality in patients with chronic lymphocytic leukemia: a joint study by ERIC, the European Research Initiative on CLL, and CLL Campus. Leukemia 2020; 34:2354.
  110. Chong EA, Roeker LE, Shadman M, et al. BTK Inhibitors in Cancer Patients with COVID-19: "The Winner Will be the One Who Controls That Chaos" (Napoleon Bonaparte). Clin Cancer Res 2020; 26:3514.
  111. Shulman RM, Weinberg DS, Ross EA, et al. Adverse Events Reported by Patients With Cancer After Administration of a 2-Dose mRNA COVID-19 Vaccine. J Natl Compr Canc Netw 2022; 20:160.
  112. Wu JT, La J, Branch-Elliman W, et al. Association of COVID-19 Vaccination With SARS-CoV-2 Infection in Patients With Cancer: A US Nationwide Veterans Affairs Study. JAMA Oncol 2022; 8:281.
  113. Cortellini A, Tabernero J, Mukherjee U, et al. SARS-CoV-2 omicron (B.1.1.529)-related COVID-19 sequelae in vaccinated and unvaccinated patients with cancer: results from the OnCovid registry. Lancet Oncol 2023; 24:335.
  114. Tan WC, Tan JYJ, Lim JSJ, et al. COVID-19 Severity and Waning Immunity After up to 4 mRNA Vaccine Doses in 73 608 Patients With Cancer and 621 475 Matched Controls in Singapore: A Nationwide Cohort Study. JAMA Oncol 2023; 9:1221.
  115. Thakkar A, Gonzalez-Lugo JD, Goradia N, et al. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell 2021; 39:1.
  116. Greenberger LM, Saltzman LA, Senefeld JW, et al. Antibody response to SARS-CoV-2 vaccines in patients with hematologic malignancies. Cancer Cell 2021; 39:1031.
  117. Ollila TA, Lu S, Masel R, et al. Antibody Response to COVID-19 Vaccination in Adults With Hematologic Malignant Disease. JAMA Oncol 2021; 7:1714.
  118. Perry C, Luttwak E, Balaban R, et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with B-cell non-Hodgkin lymphoma. Blood Adv 2021; 5:3053.
  119. Mair MJ, Berger JM, Berghoff AS, et al. Humoral Immune Response in Hematooncological Patients and Health Care Workers Who Received SARS-CoV-2 Vaccinations. JAMA Oncol 2022; 8:106.
  120. Naranbhai V, Pernat CA, Gavralidis A, et al. Immunogenicity and Reactogenicity of SARS-CoV-2 Vaccines in Patients With Cancer: The CANVAX Cohort Study. J Clin Oncol 2022; 40:12.
  121. Monin L, Laing AG, Muñoz-Ruiz M, et al. 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.
  122. Thakkar A, Gonzalez-Lugo JD, Goradia N, et al. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell 2021; 39:1081.
  123. Goshen-Lago T, Waldhorn I, Holland R, et al. Serologic Status and Toxic Effects of the SARS-CoV-2 BNT162b2 Vaccine in Patients Undergoing Treatment for Cancer. JAMA Oncol 2021; 7:1507.
  124. Eliakim-Raz N, Massarweh A, Stemmer A, Stemmer SM. Durability of Response to SARS-CoV-2 BNT162b2 Vaccination in Patients on Active Anticancer Treatment. JAMA Oncol 2021; 7:1716.
  125. Di Noia V, Pimpinelli F, Renna D, et al. Immunogenicity and Safety of COVID-19 Vaccine BNT162b2 for Patients with Solid Cancer: A Large Cohort Prospective Study from a Single Institution. Clin Cancer Res 2021; 27:6815.
  126. Fendler A, Shepherd STC, Au L, et al. Immune responses following third COVID-19 vaccination are reduced in patients with hematological malignancies compared to patients with solid cancer. Cancer Cell 2022; 40:114.
  127. Fendler A, Shepherd STC, Au L, et al. 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.
  128. Nelli F, Fabbri A, Onorato A, et al. Effects of active cancer treatment on safety and immunogenicity of COVID-19 mRNA-BNT162b2 vaccine: preliminary results from the prospective observational Vax-On study. Ann Oncol 2022; 33:107.
  129. Schmidt AL, Labaki C, Hsu CY, et al. COVID-19 vaccination and breakthrough infections in patients with cancer. Ann Oncol 2022; 33:340.
  130. Corti C, Antonarelli G, Scotté F, et al. Seroconversion rate after vaccination against COVID-19 in patients with cancer-a systematic review. Ann Oncol 2022; 33:158.
  131. Song Q, Bates B, Shao YR, et al. Risk and Outcome of Breakthrough COVID-19 Infections in Vaccinated Patients With Cancer: Real-World Evidence From the National COVID Cohort Collaborative. J Clin Oncol 2022; 40:1414.
  132. Lee LYW, Starkey T, Ionescu MC, et al. 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.
  133. Wang W, Kaelber DC, Xu R, Berger NA. Breakthrough SARS-CoV-2 Infections, Hospitalizations, and Mortality in Vaccinated Patients With Cancer in the US Between December 2020 and November 2021. JAMA Oncol 2022; 8:1027.
  134. Lim SH, Stuart B, Joseph-Pietras D, et al. Immune responses against SARS-CoV-2 variants after two and three doses of vaccine in B-cell malignancies: UK PROSECO study. Nat Cancer 2022; 3:552.
  135. Obeid M, Suffiotti M, Pellaton C, et al. Humoral Responses Against Variants of Concern by COVID-19 mRNA Vaccines in Immunocompromised Patients. JAMA Oncol 2022; 8:e220446.
  136. Valanparambil RM, Carlisle J, Linderman SL, et al. Antibody Response to COVID-19 mRNA Vaccine in Patients With Lung Cancer After Primary Immunization and Booster: Reactivity to the SARS-CoV-2 WT Virus and Omicron Variant. J Clin Oncol 2022; 40:3808.
  137. Ollila TA, Masel RH, Reagan JL, et al. Seroconversion and outcomes after initial and booster COVID-19 vaccination in adults with hematologic malignancies. Cancer 2022; 128:3319.
  138. Kim J, Chang E, Park SY, et al. Evaluation of Seropositivity After Standard Doses of Vaccination Against SARS-CoV-2 in Patients With Early Breast Cancer Receiving Adjuvant Treatment. Oncologist 2022; 27:e931.
  139. Lee LYW, Tilby M, Starkey T, et al. Association of SARS-CoV-2 Spike Protein Antibody Vaccine Response With Infection Severity in Patients With Cancer: A National COVID Cancer Cross-sectional Evaluation. JAMA Oncol 2023; 9:188.
  140. Shroff RT, Chalasani P, Wei R, et al. Immune responses to two and three doses of the BNT162b2 mRNA vaccine in adults with solid tumors. Nat Med 2021; 27:2002.
  141. Mair MJ, Mitterer M, Gattinger P, et al. Inhibition of SARS-CoV-2 Omicron BA.1 and BA.4 Variants After Fourth Vaccination or Tixagevimab and Cilgavimab Administration in Patients With Cancer. JAMA Oncol 2022; 8:1694.
  142. Rottenberg Y, Grinshpun A, Ben-Dov IZ, et al. Assessment of Response to a Third Dose of the SARS-CoV-2 BNT162b2 mRNA Vaccine in Patients With Solid Tumors Undergoing Active Treatment. JAMA Oncol 2022; 8:300.
  143. Ligumsky H, Dor H, Etan T, et al. Immunogenicity and safety of BNT162b2 mRNA vaccine booster in actively treated patients with cancer. Lancet Oncol 2022; 23:193.
  144. Gounant V, Ferré VM, Soussi G, et al. Efficacy of Severe Acute Respiratory Syndrome Coronavirus-2 Vaccine in Patients With Thoracic Cancer: A Prospective Study Supporting a Third Dose in Patients With Minimal Serologic Response After Two Vaccine Doses. J Thorac Oncol 2022; 17:239.
  145. Figueiredo JC, Ihenacho U, Merin NM, et al. SARS-CoV-2 vaccine uptake, perspectives, and adverse reactions following vaccination in patients with cancer undergoing treatment. Ann Oncol 2022; 33:109.
  146. Nelli F, Fabbri A, Onorato A, et al. Six month immunogenicity of COVID-19 mRNA-BNT162b2 vaccine in actively treated cancer patients: updated results of the Vax-On study. Ann Oncol 2022; 33:352.
  147. Fenioux C, Teixeira L, Fourati S, et al. SARS-CoV-2 Antibody Response to 2 or 3 Doses of the BNT162b2 Vaccine in Patients Treated With Anticancer Agents. JAMA Oncol 2022; 8:612.
  148. Waissengrin B, Agbarya A, Safadi E, et al. Short-term safety of the BNT162b2 mRNA COVID-19 vaccine in patients with cancer treated with immune checkpoint inhibitors. Lancet Oncol 2021; 22:581.
  149. Lasagna A, Bergami F, Lilleri D, et al. Humoral and cellular response before and after the fourth BNT162b2 vaccine dose in patients with solid tumors on active treatment. Ann Oncol 2022; 33:1207.
  150. Recommendations of the National Comprehensive Cancer Network (NCCN) Advisory Committee on COVID-19 Vaccination and Pre-exposure Prophylaxis. Available at: https://www.nccn.org/docs/default-source/covid-19/2021_covid-19_vaccination_guidance_v4-0.pdf?sfvrsn=b483da2b_66 (Accessed on December 01, 2021).
  151. ASH-ASTCT COVID-19 Vaccination for HCT and CAR T Cell Recipients: Frequently Asked Questions. Available at: https://www.hematology.org/covid-19/ash-astct-covid-19-vaccination-for-hct-and-car-t-cell-recipients (Accessed on December 01, 2021).
  152. American College of Rheumatology. COVID-19 Vaccine Clinical Guidance Summary for Patients with Rheumatic and Musculoskeletal Diseases. Available at: https://www.rheumatology.org/Portals/0/Files/COVID-19-Vaccine-Clinical-Guidance-Rheumatic-Diseases-Summary.pdf (Accessed on March 04, 2021).
  153. Garreffa E, Hamad A. Regional lymphadenopathy following COVID-19 vaccination. Eur J Cancer 2021.
  154. Skawran S, Gennari AG, Dittli M, et al. [18F]FDG uptake of axillary lymph nodes after COVID-19 vaccination in oncological PET/CT: frequency, intensity, and potential clinical impact. Eur Radiol 2022; 32:508.
  155. Lane DL, Neelapu SS, Xu G, Weaver O. COVID-19 Vaccine-Related Axillary and Cervical Lymphadenopathy in Patients with Current or Prior Breast Cancer and Other Malignancies: Cross-Sectional Imaging Findings on MRI, CT, and PET-CT. Korean J Radiol 2021; 22:1938.
  156. Editorial Comment: Prolonged Resolution of COVID-19 Vaccine–Related Axillary Lymphadenopathy Necessitates a Long Imaging Follow-Up Interval. American Journal of Roentgenology. Available at: https://www.ajronline.org/doi/full/10.2214/AJR.22.28019?utm_campaign=roentgen+journal&utm_medium=email&utm_source=informz (Accessed on November 03, 2022).
  157. Local reactions, systemic reactions, adverse events, and serious adverse events: Pfizer-BioNTech COVID-19 Vaccine. Centers for Disease Control and Prevention. Available at: https://www.cdc.gov/vaccines/covid-19/info-by-product/pfizer/reactogenicity.html (Accessed on February 25, 2021).
  158. Garreffa E, Hamad A, O'Sullivan CC, et al. Regional lymphadenopathy following COVID-19 vaccination: Literature review and considerations for patient management in breast cancer care. Eur J Cancer 2021; 159:38.
  159. Oosting SF, van der Veldt AAM, GeurtsvanKessel CH, et al. 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.
  160. Widman AJ, Cohen B, Park V, et al. Immune-Related Adverse Events Among COVID-19-Vaccinated Patients With Cancer Receiving Immune Checkpoint Blockade. J Natl Compr Canc Netw 2022; 20:1134.
  161. Ippolito E, Fiore M, Greco C, et al. COVID-19 and radiation induced pneumonitis: Overlapping clinical features of different diseases. Radiother Oncol 2020; 148:201.
  162. Berlin A, Lovas M, Truong T, et al. Implementation and Outcomes of Virtual Care Across a Tertiary Cancer Center During COVID-19. JAMA Oncol 2021; 7:597.
  163. Viers BR, Lightner DJ, Rivera ME, et al. Efficiency, satisfaction, and costs for remote video visits following radical prostatectomy: a randomized controlled trial. Eur Urol 2015; 68:729.
  164. Darcourt JG, Aparicio K, Dorsey PM, et al. Analysis of the Implementation of Telehealth Visits for Care of Patients With Cancer in Houston During the COVID-19 Pandemic. JCO Oncol Pract 2021; 17:e36.
  165. Asan O, Cooper Ii F, Nagavally S, et al. Preferences for Health Information Technologies Among US Adults: Analysis of the Health Information National Trends Survey. J Med Internet Res 2018; 20:e277.
  166. Eberly LA, Kallan MJ, Julien HM, et al. Patient Characteristics Associated With Telemedicine Access for Primary and Specialty Ambulatory Care During the COVID-19 Pandemic. JAMA Netw Open 2020; 3:e2031640.
  167. Hsiehchen D, Muquith M, Haque W, et al. Clinical Efficiency and Safety Outcomes of Virtual Care for Oncology Patients During the COVID-19 Pandemic. JCO Oncol Pract 2021; 17:e1327.
  168. Breen KE, Tuman M, Bertelsen CE, et al. Factors Influencing Patient Preferences for Telehealth Cancer Genetic Counseling During the COVID-19 Pandemic. JCO Oncol Pract 2022; 18:e462.
  169. Arem H, Moses J, Cisneros C, et al. Cancer Provider and Survivor Experiences With Telehealth During the COVID-19 Pandemic. JCO Oncol Pract 2022; 18:e452.
  170. Parrinello G, Missale F, Sampieri C, et al. Safe management of laryngectomized patients during the COVID-19 pandemic. Oral Oncol 2020; 107:104742.
  171. Brook I. Prevention of COVID-19 Infection in Neck Breathers, Including Laryngectomees. Int Arch Otorhinolaryngol 2020; 24:e253.
  172. Varghese JJ, Aithal VU, Rajashekhar B. Self-care and clinical management of persons with laryngectomy during COVID-19 pandemic: a narrative review. Support Care Cancer 2021; 29:7183.
  173. Gallo O, Trotta M, Orlando P, et al. SARS-CoV-2 in upper and lower airway samples of a laryngectomized patient: New insights and many lessons. Oral Oncol 2020; 107:104841.
  174. Patel TR, Teitcher JE, Tajudeen BA, Revenaugh PC. Disparate Nasopharyngeal and Tracheal COVID-19 Diagnostic Test Results in a Patient With a Total Laryngectomy. Otolaryngol Head Neck Surg 2020; 163:710.
Topic 128993 Version 78.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟