Next Article in Journal
Vaccine-Related Autoimmune Hepatitis: Emerging Association with SARS-CoV-2 Vaccination or Coincidence?
Previous Article in Journal
Monkeypox Disease (MPOX) Perceptions among Healthcare Workers versus General Population during the First Month of the WHO Alert: Cross-Sectional Survey in Saudi Arabia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Impact of COVID-19 on Cancer Care: An Update

1
Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, India
2
Department of Reproductive Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
3
Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA
*
Authors to whom correspondence should be addressed.
Vaccines 2022, 10(12), 2072; https://doi.org/10.3390/vaccines10122072
Submission received: 23 October 2022 / Revised: 26 November 2022 / Accepted: 28 November 2022 / Published: 4 December 2022
(This article belongs to the Section Clinical Immunology)

Abstract

:
The world has been affected socioeconomically for the last two years due to the emergence of different variants of the COVID-19 virus. Vaccination is the major and most efficient way to prevent the widening of this pandemic. Those who are having comorbidities are more vulnerable to serious infections due to their immunocompromised state. Additionally, cancer patients could be at significant risk for COVID-19. In this pandemic era, the diagnosis and treatment of cancer were significantly affected. Clinical trials at the initial stage were performed on healthy or COVID-19 infected patients. This produces a greater level of hesitancy in cancer patients. This review article provide an update regarding the vaccination and treatment for COVID-19 in patients with cancer and future directions.

1. Introduction

The COVID-19 outbreak has caused catastrophe in health care systems, raising worries about its influence on non-COVID illnesses [1,2]. Cancer diagnosis and treatment are time-sensitive, and any delay is likely to have serious consequences for patients. Since no preparation strategy can completely defend against black-swan occurrences like the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak, it is crucial to guarantee that such incidents do not lead to the collapse of critical healthcare sectors. During the COVID-19 pandemic, the World Health Organization (WHO) conducted a Global Pulse Survey that looked at the continuity of essential health services. According to the report, between 5 and 50% of cancer care-screening and treatment was disrupted in all countries as reported in the last quarter of 2021 [3]. This is despite the improvement since the first quarter of last year, in which service disruptions exceeded 50% in 44% of countries, and ranged between 5 and 50% in the rest [3]. The complex and delayed reaction to COVID-19 serves as a wake-up call to governments throughout the world that healthcare must be emphasized worldwide, and that affordable healthcare might be a fundamental right that cannot be neglected [4]. Almost 3–4 percent of all COVID-19 victims have cancer and related diseases, and the numbers are terrifying, as the patient is described as “doubly unlucky” to be suffering from cancer as well as being infected with COVID-19. Cancer patients could be at significant risk for COVID-19, and there seems to be an upsurge in the likelihood of serious conditions, such as ICU usage and artificial ventilation, for them as opposed to non-cancer COVID-19 patients [5]. Other than cancer, people who are suffering from medical conditions such as cardiovascular disease, diabetes, and, chronic respiratory disease are also more likely to develop serious illnesses [6]. The immunocompromised status of patients is almost certainly the reason for this elevated danger to cancer victims with COVID-19. The lethal combination of COVID-19 and cancer is also being blamed on cytokine storms. Patients with cancer and COVID-19 both have inflammatory responses [7]. Because the inflammation is pervasive, the misery of cancer patients is exacerbated. Additionally, many molecular and biological processes are also reported in patients with cancer, such as a cytokine storm, increased angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) production, in addition to coagulopathy [8], which is a potential concern observed in a lot of cancer patients (Figure 1). In respiratory system cells, the increased amount of Anexelekto (AXL) is also reported. It plays an important role in cancers and in COVID-19 as a transmembrane protein that promotes cell growth, migration, aggregation, metastasis, and adhesion [9]. The management and cure of cancer is handled by many nanotechnology based products. [10,11,12]. Herbal drugs are converted to nano pharmaceuticals to provide effective cancer management [13,14,15].
This review aims to highlight the requirement for substantial clinical proof of the efficacy and safety profile of several COVID-19 vaccines in order to motivate patients to get immunized. To give the best care, the impact of SARS-CoV-2 on the different forms of cancer differs and has to be thoroughly researched. The review makes the claim that it will help readers comprehend how cancer and other co-occurring disorders are linked to COVID. It also focuses on the hesitancy, its cause, and the necessary precautions to be undertaken before receiving a vaccine.

2. Cancer Diagnostics and COVID-19

According to Allegra et al. [17], COVID-19 and carcinoma have similar markers such as the carbohydrate antigens (CA) 125 and 153, carcinoembryonic antigen (CEA), human epididymis protein 4 (HE4), C-reactive protein (CRP), and cytokeratin-19 fragment (CYFRA21-1); However, establishing whether the increase in such biomarkers is the result of COVID-19, malignancy, or both at the same time is a challenge for hospitals and clinicians (Figure 1) [14]. The importance of diagnosis cannot be overstated, and it necessitates broad information on the pathophysiology of the lethal pair of COVID-19 and malignancy. According to one study, the broad impact of COVID-19 on cancer patients and the system that assists them may cause mortality among cancer patients in the United Kingdom to increase by 20% over the course of the following 12 months, resulting in 6000 more deaths. [18]. As per Englum et al. [19], There are 4.1 million cancer-related interactions, 3.9 million applicable operations, and 251,647 new malignancies diagnosed between 2018 and 2020. In 2020, colonoscopies declined by 45 percent compared to the yearly averages from 2018 to 2019, while prostate biopsies, chest computed tomography scans, and cystoscopies decreased by 29%, 10%, and 21 %, respectively [13]. The number of new cancer diagnoses fell from 13% to 23%. Despite decreases in pandemic-related limitations, these declines varied by region and continued to accrue. Therefore, “Post-pandemic cancer diagnoses across races/ethnicities should be tracked to see whether measures to boost equality in cancer detection and early treatment are bearing fruit”, noted Caswell-Jin [20]. In one another study, a total of 10,404 participants in the cohort of 23,266 cancer patients and 1,784,293 cancer-free participants reported shows results like a positive COVID-19 test. Cancer survivors had a 60% increased risk of a positive COVID-19 test compared to those without cancer. The odds of a positive test were 2.2-fold higher among patients with cancer receiving chemotherapy or immunotherapy. Among participants over 65 years of age and males, COVID-19 infection was associated with a higher risk of cancer [21]. In the United States, cancer-related mortality rose by 3% during the pandemic era, most likely as a result of delayed diagnoses and inefficient patient care that forces hospitals to treat patients at high risk of contracting COVID-19. According to the retrospective research of two institutes, a positive COVID-19 test caused a delay in the length of cancer therapy. The length of the delays varies depending on a number of factors, including time between test results, a string of positive tests, fear of medical treatment and hospitalization, etc. A total of 131 cancer patients who were infected with COVID required hospitalization, of whom 38% required ICU care, and 7% died [22].

3. COVID-19 and Cancer Treatment

Cancer patients are more likely to be hospitalized and die after contracting SARS-CoV-2 [23]. As a result, we sought to undertake one of the first population-level analyses of vaccination efficacy against SARS-CoV-2 outbreaks in cancer patients [24]. Chemotherapy and immune suppressants used after surgical cancer excision typically damage the patient’s immune system. This implies that COVID-19 may harm any such patient more than it would a healthy person. Children who get anti-cancer medication and hematopoietic stem cell transplantation (HSCT) have an increased risk of serious complications (up to 20%) and death (4–5%). The majority of these data come from the registry or institution-based sources and predate the Delta/Omicron variants’ dominance [25]. The therapy must be carried out while keeping the extra COVID-19 risk factors connected with cancer in mind. Certain anti-cancer therapies may necessitate a reduction in immunity to destroy cancer cells that provide a fair chance of SARS-CoV-2 infecting such patients. To prevent patients from experiencing the lethal combination of COVID-19 and cancer further, physicians and cancer organizations recommend delaying cytotoxic treatment until COVID negative diagnostic results in such infected patients [26,27,28].
Human tissues other than the lungs express TMPRSS2 and ACE2, including the testes, which are susceptible to SARS-CoV-2 infection. A very common alteration in prostate cancer is the fusion of TMPRSS2 with another androgen-responsive gene. In their study, Bhowmick NA et al. emphasized the evidence that TMPRSS2 and ACE2 are affected by androgens, as well as the implications of using androgen suppression to inhibit TMPRSS2 to target SARS-CoV-2 [29].
Cancer treatment is primarily provided by pre-pandemic standards in regions with a low prevalence of viral infection. It is often advised that treatment be postponed until it is certain the patient will not get COVID-19 when a patient is known to have been infected with SARS-CoV-2, especially if he or she has not had a COVID-19 vaccination or if the vaccine is anticipated to have an unsatisfactory response. Patients undergoing low-risk cancer medicines, such as hormone medications, may be exempt from receiving therapy [30]. For patients with low-viral transmission systemic therapy, radiation therapy and surgery have been undertaken [31].
Cancer treatment in locations with strong viral transmission-the parts that follow will focus on the challenges associated with systemic anticancer medicines, radiation therapy, and surgery in regions with high rates of persistent viral transmission during the pandemic’s peak times of activity. In areas where the virus is less prevalent, all of these characteristics are less significant [32].
Barrière et al. provide five suggestions to health authorities and healthcare professionals [33].
Statement 1: All cancer patients should have a third vaccination dose three to four months after receiving the second dose (Dose 2), along with controls for any remaining anti-S serum antibodies, as cancer patients have a lower median anti-S antibody titer after two vaccine doses than the general population. The data currently available demonstrate quicker Omicron VOC immunity fading than the Delta strain, but enhanced protection following booster dosing (Dose 3) [34].
Statement 2: Decide that if a patient’s serum anti-SARS-CoV-2 S (antiS) titer falls below 1000 BAU/mL, the fourth dose of vaccination should be suggested. This cut-off corresponds to titers seen in a group of individuals without comorbidities 3–4 months after Dose 2, for whom Dose 3 treatment is currently recommended in the majority of industrialized countries to give the best protection against the Omicron variant.
Statement 3: Monoclonal antibody (mAB) tixagevimab/cilgavimab (EVUSHELD, AstraZeneca) is used for pre-exposure prophylaxis for cancer patients who do not have seroconversion after Dose 3 or 4. This combination of long-acting anti-S mAbs has a remarkable effect on in vitro efficacy of Delta VOC and is less affected by several Omicron S mutations [33,35].
Statement 4-A three-dose complete vaccination for the patient’s family should be suggested as soon as 4 months following Dose 2. Additionally, when in close, frequent contact with immune-compromised parents, the majority of Scientific Pediatric Societies, as well as the US (Food and Drug Administration) and European Medicines Agency (EMA) Health Agency advise the immunizing children between the ages of 5 and 11 [36].
Statement 5: Patients should use a high-protection mask that filters at least 95% of particles(N95) and has a filtering facepiece type 2 (FFP2). Such masks should be required in crowded environments, whether on public or private transport (when shared with others). Recent comparative data demonstrates that these masks offer more effective personal protection than surgical masks [37].

4. COVID-19 Vaccine and Cancer

According to current prime-boost theories, patients with cancer can receive the COVID-19 vaccine in a safe and therapeutically effective way [38,39,40]. The COVID-19 vaccines are considered efficacious against the majority of viral variants, such as the Delta form. Variants may, nevertheless, promote disease in certain vaccinated persons. The Omicron form, for example, is more pathogenic than the Delta variant and may produce recurrent infections in vaccinated patients. Booster dosages are critical in lowering the chance of a reoccurring illness [41,42,43]. Slomski reported [44] that the trial, which was undertaken with the Pfizer COVID-19 vaccine in Israel, included 232 cancer patients along with a control group of 261 healthcare workers. The results after the administration of the first immunization are average, but after the second vaccine, almost 86% of the cancer patients demonstrated sufficient neutralizing antibodies against SARS-CoV-2. Experts agree that COVID-19 immunization is appropriate for cancer patients, survivors, and those receiving cancer treatment, including chemoimmunotherapy [45]. The best current information shows that if you have cancer, your chances of dying or having serious problems with COVID-19 are approximately two times higher than those with no cancer. Until now, patients receiving cancer therapies had been precluded from COVID-19 vaccination studies. Our presumptions are thus based on what we understand about the safety and effectiveness of the finest vaccine candidates, the assessment of conventional vaccination in patients with cancer, and the immunological alterations brought on by current cancer treatments. To increase the effectiveness of the vaccine in immunocompromised cancer patients, nanotechnology must be used as a means of building a successful vaccine platform that can be manufactured in a scalable manner and distributed worldwide as well as delivered faster and more precisely [46]. The study team found that 79% of vaccine-treated cancer patients displayed a T-cell response, and comparable findings were observed in patients with solid tumors and blood cancers. It is noteworthy that even with the delta version, the vaccines were seen to activate T cells in those who had not developed a neutralizing antibody response by the time they received the second dose [47]. According to the findings of Obeid et al. [48], with a comparative efficacy study, there are neutralizing antibodies (nAbs) after two doses of vaccine in 50% of a cancer patients that seems to lose the protection after six months of immunization. These findings might aid in the development of the optimum boosting immunization regimen, particularly for immunocompromised individuals. Fendler et al. [49] conducted CAPTURE research and demonstrated that “Previous SARS-CoV-2 infection boosted the nAb response including against VOC, and anti-CD20 treatment was associated with undetectable nAbT. Vaccine-induced T cell responses were detected in 80% of patients and were comparable between vaccines or cancer types”.
New observational research published by Sheeba Irshad et al. involved 151 cancer patients, including 95 with organ cancer and 56 patients with hematological cancer [50]. There were no documented vaccine-related fatalities. One administration of the BNT162b2 vaccination is ineffective in cancer patients. Immunogenicity was dramatically enhanced in patients with solid tumors after 2 weeks of a vaccination boost on day 21 following the first dose. These findings support the prioritizing of cancer patients for an early (day 21) second BNT162b2 vaccination. According to a new study, the Omicron variant of SARS-CoV-2 is more hazardous in cancer patients than in the general population. Despite this, the death rate among cancer patients infected with omicron is lower than in prior SARS-CoV-2 variants [51]. More research focusing on strategies to boost vaccination efficacy and the identification of novel treatment options is critically required for these patients. Three months following the second immunization dose (Dose 2), all cancer patients should get a third dose of the vaccine, along with any further serum anti-S antibody controls [36].

5. Vaccines and Their Ongoing Clinical Trials

An international panel of oncology experts examined the long-term consequences of the pandemic on cancer clinical trials using new data on worldwide clinical trials gathered by IQVIA, and suggests remedies for clinical trial stakeholders [52]. Oncology trials may be time-consuming, costly, and stressful for patients and their families, complicating the decision to enroll even before concerns about COVID-19 arose [53].
Patients who have had malignant tumors may have diminished immunity as a result of immunosenescence, previous treatment, or comorbidities, which will affect the effectiveness of COVID-19 vaccines [54]. People with active cancer and people taking immunosuppressive medicines, however, were excluded from some vaccination trials, which questions their effectiveness and safety [18]. A significant portion of participants in the randomized, placebo-controlled, observer-blinded phase 1/2/3 BNT162b2 mRNA COVID-19 vaccination trial had a history of neoplasm at baseline, either past or present [55]. This study will also offer a post hoc subgroup analysis of clinical efficacy and safety in persons with a history of neoplasm from this experiment in response to the need for solid confirmation of vaccination effectiveness and toxicity in cancer patients [56]. Our post hoc subgroup analysis of the clinical efficacy and safety of this vaccine in individuals who have a history of malignancy fulfills the requirement for direct proof of vaccination efficacy and safety in cancer patients [57]. BNT162b2 was 95% effective in preventing COVID-19 in people 16 years of age from 7 days post-dose two after a median follow-up of 2 months in the overall study group. Later studies in teenagers (aged 12 to 15) revealed 100% efficacy and non-inferior immunogenicity compared to young adults (aged 16 to 25) [58]. For dose two, the updated vaccination efficacy throughout the whole trial cohort was still high (91%) after up to 6 months of follow-up. Based on these findings, the US FDA granted emergency use authorization for the BNT162b2 COVID-19 vaccine in December 2020, and the European Union granted conditional marketing clearance for the immunization of individuals 12 years of age and older [59]. Recently, mRNA-1273 or BNT162b2 mRNA vaccines have been reported to increase resistance to neutralizing antibodies of the SARS-CoV-2 Omicron variant. However, an additional, third dose of the mRNA booster vaccine appears to provide much stronger protection against Omicron, similar to what has been reported for other variants of concern [60]. A patient with cancer who develops breakthrough COVID-19 following a full vaccination remains susceptible to severe outcomes [61]. On 23 August 2021, the FDA approved the BNT162b2 COVID-19 vaccine after receiving a biologics license application for individuals 16 years of age and older. Trial participants who were not using immunosuppressive drugs at baseline but had a prior prognosis of any cancer or other neoplasm were included in the current post hoc subgroup analysis [62]. Being almost neglected during the initial trials of the COVID-19 vaccine, healthcare is healthcare is now focusing on the immunosuppressed patients as well. Cancer patients who have received two doses of the mRNA vaccine (Spikevax and Comirnaty) are the subjects of recruitment research. The focus of the study is on how cancer patients’ humoral immunity develops (NCT05075538). Patients with solid tumors, multiple myeloma, and hematologic malignancy were included in a phase-2 study of mRNA-1273, which demonstrated the drug’s immunogenicity and safety profile (NCT04847050). On the patient with malignant solid neoplasm, another observational study is carried out to determine the safety and effectiveness of the vaccination. Its major consequence, antibody production, and t-cell-mediated immunization are the outcome criteria. The investigation should be finished by 31 December 2023. (NCT04865133). The long-term safety of patients who will be subjected to therapy that might lower their immunity is a serious worry. As COVID-19 targets lung cancer, the situation there might deteriorate. An observational trial of 300 lung cancer patients focused on the long-term impact and safety profile of the vaccination (NCT04894682). Immunotherapy is one of the major approaches for the treatment of cancer. A study on 53 patients was performed to evaluate the immune response produced in cancer patients exposed to immunotherapy and normal cancer patients (NCT05062525).
Vaccine hesitancy is more prevalent in malignant patients as compared to the normal individual. A study conducted on 429 patients concluded that 48% of the population was unsure regarding the vaccine consumption primarily due to safety concerns (32%) and about the complication it might cause (12%) [63]. According to a different study on leukemia patients and survivors, 17% of all responders were unlikely to receive the vaccination. The dread of their health condition getting worse and worries about the safety of the vaccination were two potential causes [46,64].

6. Discussion

A cancer patient receives many treatments, the bulk of which target the immune system. According to a research, cancer patients have a greater fatality rate and need for intensive care than healthy people do. Immunosuppression in patients limits the use of live vaccines due to an increased risk of infection. However, immunization is necessary to guard against novel variations. If the patient is commencing chemotherapy, they should allow at least 14 days before the immunization. A patient with blood cancer who is receiving chemotherapy should not be vaccinated until the neutrophils achieve homeostasis. This circumstance has caused cancer therapy to be postponed or interrupted. This element may make the malignancy worse and, in extreme situations, may be lethal. According to clinical research on 18 cancer patients who had recently contracted COVID-19, the fatality rate for cancer patients had increased to 5.6% from 2.3%. This emphasizes that in order to offer proper care and maintain patient safety, a thorough investigation regarding the effects of real illness and vaccination must be conducted.

7. Concluding Remarks

The COVID-19 pandemic encourages us to develop viable strategies for dealing with subsequent health crises [65,66,67,68,69,70]. However, there was some data redundancy or duplication in tests and experiments due to the heterogeneity in inclusion criteria between distinct cancer registries that were restricted to different subgroups or regions. Going forward, coordinated standardized initiatives in which various registries collaborate to build a uniform method for consistent data collecting and reporting could yield speedier, more precise, and more robust clinical implementation outcomes. More research is required to prove the safety and efficacy of therapeutic molecules in COVID-19 with comorbidities such as cancer. Future medical science demands a better understanding of various drug-drug and drug-pathological interaction and develops preventive measures to ensure the wellbeing of the patient.

Author Contributions

Conceptualization, V.P.C., V.P.C., F.-F.P. and Z.-S.C. have written the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

We thank the Six Talent Peaks of Jiangsu Province (YY-128), and the Wuxi Taihu Lake Talent Plan Top Talents Projects (BJ 2020001) for their support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

V.P.C. wants to dedicate this work to the L. M. College of Pharmacy as a part of the 75th anniversary of the college. Figure 1 is created with Biorender.com. V.P.C. wants to thanks Pankti Balar and Aayushi Patel for her support in manuscript formatting.

Conflicts of Interest

The authors declare no conflict of interest concerning the authorship and publication of this article.

References

  1. Chavda, V.P.; Feehan, J.; Apostolopoulos, V. A Veterinary Vaccine for SARS-CoV-2: The First COVID-19 Vaccine for Animals. Vaccines 2021, 9, 631. [Google Scholar] [CrossRef] [PubMed]
  2. Chavda, V.P.; Kapadia, C.; Soni, S.; Prajapati, R.; Chauhan, S.C.; Yallapu, M.M.; Apostolopoulos, V. A Global Picture: Therapeutic Perspectives for COVID-19. Immunotherapy 2022, 14, 351–371. [Google Scholar] [CrossRef] [PubMed]
  3. Cancer Services Disrupted by up to 50% in All Countries Reporting: A Deadly Impact of COVID-19. Available online: https://www.who.int/europe/news/item/03-02-2022-cancer-services-disrupted-by-up-to-50-in-all-countries-reporting-a-deadly-impact-of-covid-19 (accessed on 24 September 2022).
  4. Pramesh, C.S.; Chinnaswamy, G.; Sengar, M.; Ranganathan, P.; Badwe, R. COVID-19 and Cancer Care in India. Nat. Cancer 2021, 2, 1257–1259. [Google Scholar] [CrossRef] [PubMed]
  5. Dai, M.; Liu, D.; Liu, M.; Zhou, F.; Li, G.; Chen, Z.; Zhang, Z.; You, H.; Wu, M.; Zheng, Q.; et al. Patients with Cancer Appear More Vulnerable to SARS-CoV-2: A Multicenter Study during the COVID-19 Outbreak. Cancer Discov. 2020, 10, 783–791. [Google Scholar] [CrossRef] [PubMed]
  6. Coronavirus. Available online: https://www.who.int/health-topics/coronavirus (accessed on 24 September 2022).
  7. Sinha, S.; Kundu, C.N. Cancer and COVID-19: Why Are Cancer Patients More Susceptible to COVID-19? Med. Oncol. 2021, 38, 101. [Google Scholar] [CrossRef] [PubMed]
  8. Farahani, M.; Niknam, Z.; Mohammadi Amirabad, L.; Amiri-Dashatan, N.; Koushki, M.; Nemati, M.; Danesh Pouya, F.; Rezaei-Tavirani, M.; Rasmi, Y.; Tayebi, L. Molecular Pathways Involved in COVID-19 and Potential Pathway-Based Therapeutic Targets. Biomed. Pharm. 2022, 145, 112420. [Google Scholar] [CrossRef]
  9. Zhang, W.-N.; Li, X.-P.; Wang, P.-F.; Zhu, L.; Xiao, X.-H.; Dai, Y.-J. Comprehensive Analysis of the Novel Omicron Receptor AXL in Cancers. Comput. Struct. Biotechnol. J. 2022, 20, 3304–3312. [Google Scholar] [CrossRef]
  10. Chavda, V.P. Nanotherapeutics and Nanobiotechnology. In Applications of Targeted Nano Drugs and Delivery Systems; Elsevier: Amsterdam, The Netherlands, 2019; pp. 1–13. [Google Scholar]
  11. Chen, R.-P.; Chavda, V.P.; Patel, A.B.; Chen, Z.-S. Phytochemical Delivery Through Transferosome (Phytosome): An Advanced Transdermal Drug Delivery for Complementary Medicines. Front. Pharm. 2022, 13, 850862. [Google Scholar] [CrossRef]
  12. Chavda, V.P. Chapter 4 Nanobased Nano Drug Delivery A Comprehensive Review. In Applications of Targeted Nano Drugs and Delivery Systems; Elsevier: Amsterdam, The Netherlands, 2021; pp. 69–92. [Google Scholar]
  13. Chavda, V.P.; Vihol, D.; Mehta, B.; Shah, D.; Patel, M.; Vora, L.K.; Pereira-Silva, M.; Paiva-Santos, A.C. Phytochemical-Loaded Liposomes for Anticancer Therapy: An Updated Review. Nanomedicine 2022, 17, 547–568. [Google Scholar] [CrossRef]
  14. Chavda, V.P.; Patel, A.B.; Mistry, K.J.; Suthar, S.F.; Wu, Z.-X.; Chen, Z.-S.; Hou, K. Nano-Drug Delivery Systems Entrapping Natural Bioactive Compounds for Cancer: Recent Progress and Future Challenges. Front. Oncol. 2022, 12, 867655. [Google Scholar] [CrossRef]
  15. Chavda, V.P.; Ertas, Y.N.; Walhekar, V.; Modh, D.; Doshi, A.; Shah, N.; Anand, K.; Chhabria, M. Advanced Computational Methodologies Used in the Discovery of New Natural Anticancer Compounds. Front. Pharm. 2021, 12, 702611. [Google Scholar] [CrossRef]
  16. Bora, V.R.; Patel, B.M. The Deadly Duo of COVID-19 and Cancer! Front. Mol. Biosci. 2021, 8, 643004. [Google Scholar] [CrossRef]
  17. Allegra, A.; Pioggia, G.; Tonacci, A.; Musolino, C.; Gangemi, S. Cancer and SARS-CoV-2 Infection: Diagnostic and Therapeutic Challenges. Cancers 2020, 12, 1581. [Google Scholar] [CrossRef] [PubMed]
  18. Lai, A.G.; Pasea, L.; Banerjee, A.; Hall, G.; Denaxas, S.; Chang, W.H.; Katsoulis, M.; Williams, B.; Pillay, D.; Noursadeghi, M.; et al. Estimated Impact of the COVID-19 Pandemic on Cancer Services and Excess 1-Year Mortality in People with Cancer and Multimorbidity: Near Real-Time Data on Cancer Care, Cancer Deaths and a Population-Based Cohort Study. Bmj Open 2020, 10, e043828. [Google Scholar] [CrossRef]
  19. Englum, B.R.; Prasad, N.K.; Lake, R.E.; Mayorga-Carlin, M.; Turner, D.J.; Siddiqui, T.; Sorkin, J.D.; Lal, B.K. Impact of the COVID-19 Pandemic on Diagnosis of New Cancers: A National Multicenter Study of the Veterans Affairs Healthcare System. Cancer 2022, 128, 1048–1056. [Google Scholar] [CrossRef]
  20. Gallagher, A. Breast Cancer Diagnosis & Treatment Before vs. During COVID-19 Pandemic. Oncol. Times 2022, 44, 27. [Google Scholar]
  21. Lee, K.A.; Ma, W.; Sikavi, D.R.; Drew, D.A.; Nguyen, L.H.; Bowyer, R.C.E.; Cardoso, M.J.; Fall, T.; Freidin, M.B.; Gomez, M.; et al. Cancer and Risk of COVID-19 Through a General Community Survey. Oncologist 2021, 26, e182–e185. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, J.T.-Y.; Kwon, D.H.; Glover, M.J.; Henry, S.; Wood, D.; Rubin, D.L.; Koshkin, V.S.; Schapira, L.; Shah, S.A. Changes in Cancer Management Due to COVID-19 Illness in Patients with Cancer in Northern California. Jco Oncol. Pr. 2021, 17, e377–e385. [Google Scholar] [CrossRef]
  23. Belsky, J.A.; Tullius, B.P.; Lamb, M.G.; Sayegh, R.; Stanek, J.R.; Auletta, J.J. COVID-19 in Immunocompromised Patients: A Systematic Review of Cancer, Hematopoietic Cell and Solid Organ Transplant Patients. J. Infect. 2021, 82, 329–338. [Google Scholar] [CrossRef] [PubMed]
  24. Vaccine Effectiveness against COVID-19 Breakthrough Infections in Patients with Cancer (UKCCEP): A Population-Based Test-Negative Case-Control Study—The Lancet Oncology. Available online: https://www.thelancet.com/journals/lanonc/article/PIIS1470-2045(22)00202-9/fulltext (accessed on 24 September 2022).
  25. Liu, A.P.Y.; Lam, G.K.S.; Chan, W.Y.K.; Chow, T.T.W.; Cheung, J.; Wong, S.C.Y.; Leung, W.; Lee, P.P.W.; Cheng, F.W.T.; Chan, G.C.F. SARS-CoV-2 Infection in Children Undergoing Oncologic Treatment in Hong Kong: A Population-Based Cohort during the Omicron Wave. Pediatric Blood Cancer 2022, e29894. [Google Scholar] [CrossRef]
  26. Al-Shamsi, H.O.; Alhazzani, W.; Alhuraiji, A.; Coomes, E.A.; Chemaly, R.F.; Almuhanna, M.; Wolff, R.A.; Ibrahim, N.K.; Chua, M.L.K.; Hotte, S.J.; et al. A Practical Approach to the Management of Cancer Patients During the Novel Coronavirus Disease 2019 (COVID-19) Pandemic: An International Collaborative Group. Oncologist 2020, 25, e936–e945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Borcherding, N.; Jethava, Y.; Vikas, P. Repurposing Anti-Cancer Drugs for COVID-19 Treatment. Drug Des. Dev. 2020, 14, 5045–5058. [Google Scholar] [CrossRef]
  28. Singh, T.U.; Parida, S.; Lingaraju, M.C.; Kesavan, M.; Kumar, D.; Singh, R.K. Drug Repurposing Approach to Fight COVID-19. Pharm. Rep. Pr 2020, 72, 1479–1508. [Google Scholar] [CrossRef] [PubMed]
  29. Bhowmick, N.A.; Oft, J.; Dorff, T.; Pal, S.; Agarwal, N.; Figlin, R.A.; Posadas, E.M.; Freedland, S.J.; Gong, J. COVID-19 and Androgen-Targeted Therapy for Prostate Cancer Patients. Endocr. Relat. Cancer 2020, 27, R281–R292. [Google Scholar] [CrossRef]
  30. Sud, A.; Jones, M.E.; Broggio, J.; Loveday, C.; Torr, B.; Garrett, A.; Nicol, D.L.; Jhanji, S.; Boyce, S.A.; Gronthoud, F.; et al. Collateral Damage: The Impact on Outcomes from Cancer Surgery of the COVID-19 Pandemic. Ann. Oncol. 2020, 31, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  31. COVID-19: Considerations in Patients with Cancer—UpToDate. Available online: https://www.uptodate.com/contents/covid-19-considerations-in-patients-with-cancer#H1193654967 (accessed on 24 September 2022).
  32. Fligor, S.C.; Wang, S.; Allar, B.G.; Tsikis, S.T.; Ore, A.S.; Whitlock, A.E.; Calvillo-Ortiz, R.; Arndt, K.R.; Gangadharan, S.P.; Callery, M.P. Gastrointestinal Malignancies and the COVID-19 Pandemic: Evidence-Based Triage to Surgery. J. Gastrointest Surg. 2020, 24, 2357–2373. [Google Scholar] [CrossRef]
  33. 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. [Google Scholar] [CrossRef]
  34. Gruell, H.; Vanshylla, K.; Tober-Lau, P.; Hillus, D.; Schommers, P.; Lehmann, C.; Kurth, F.; Sander, L.E.; Klein, F. MRNA Booster Immunization Elicits Potent Neutralizing Serum Activity against the SARS-CoV-2 Omicron Variant. Nat. Med. 2022, 28, 477–480. [Google Scholar] [CrossRef]
  35. Chavda, V.P.; Prajapati, R.; Lathigara, D.; Nagar, B.; Kukadiya, J.; Redwan, E.M.; Uversky, V.N.; Kher, M.N.; Patel, R. Therapeutic Monoclonal Antibodies for COVID-19 Management: An Update. Expert Opin. Biol. Ther. 2022, 22, 763–780. [Google Scholar] [CrossRef]
  36. Barrière, J.; Zalcman, G.; Fignon, L.; Peiffer-Smadja, N.; Audigier-Valette, C.; Carles, M. Omicron Variant: A Clear and Present Danger for Patients with Cancer. Eur. J. Cancer 2022, 165, 25–26. [Google Scholar] [CrossRef]
  37. Bagheri, G.; Thiede, B.; Hejazi, B.; Schlenczek, O.; Bodenschatz, E. An Upper Bound on One-to-One Exposure to Infectious Human Respiratory Particles. Proc. Natl. Acad. Sci. USA 2021, 118, e2110117118. [Google Scholar] [CrossRef]
  38. COVID-19 Vaccines in Patients with Cancer: Immunogenicity, Efficacy and Safety|Nature Reviews Clinical Oncology. Available online: https://www.nature.com/articles/s41571-022-00610-8 (accessed on 24 September 2022).
  39. Chavda, V.P.; Yao, Q.; Vora, L.K.; Apostolopoulos, V.; Patel, C.A.; Bezbaruah, R.; Patel, A.B.; Chen, Z.-S. Fast-Track Development of Vaccines for SARS-CoV-2: The Shots That Saved the World. Front. Immunol. 2022, 13. [Google Scholar] [CrossRef] [PubMed]
  40. Chavda, V.P.; Apostolopoulos, V. COVID-19 Vaccine Design and Vaccination Strategy for Emerging Variants. Expert Rev. Vaccines 2022, 21, 1359–1361. [Google Scholar] [CrossRef] [PubMed]
  41. Chavda, V.P.; Apostolopoulos, V. Is Booster Dose Strategy Sufficient for Omicron Variant of SARS-CoV-2? Vaccines 2022, 10, 367. [Google Scholar] [CrossRef]
  42. Chavda, V.P.; Apostolopoulos, V. Global Impact of Delta plus Variant and Vaccination. Expert Rev. Vaccines 2022, 21, 597–600. [Google Scholar] [CrossRef]
  43. Chavda, V.P.; Apostolopoulos, V. Omicron Variant (B.1.1.529) of SARS-CoV-2: Threat for the Elderly? Maturitas 2022, 158, P78–P81. [Google Scholar] [CrossRef]
  44. Slomski, A. Most Fully Vaccinated Patients With Cancer Have SARS-CoV-2 Antibodies. JAMA 2021, 326, 800. [Google Scholar] [CrossRef] [PubMed]
  45. Hwang, J.K.; Zhang, T.; Wang, A.Z.; Li, Z. COVID-19 Vaccines for Patients with Cancer: Benefits Likely Outweigh Risks. J. Hematol. Oncol. 2021, 14, 38. [Google Scholar] [CrossRef]
  46. Han, H.J.; Nwagwu, C.; Anyim, O.; Ekweremadu, C.; Kim, S. COVID-19 and Cancer: From Basic Mechanisms to Vaccine Development Using Nanotechnology. Int. Immunopharmacol. 2021, 90, 107247. [Google Scholar] [CrossRef]
  47. Mahase, E. Covid-19: Just a Third of Blood Cancer Patients Had Antibodies against Delta Variant after Two Vaccine Doses, Study Finds. BMJ 2021, 375, n2623. [Google Scholar] [CrossRef]
  48. Obeid, M.; Suffiotti, M.; Pellaton, C.; Bouchaab, H.; Cairoli, A.; Salvadé, V.; Stevenel, C.; Hottinger, R.; Pythoud, C.; Coutechier, L.; et al. Humoral Responses Against Variants of Concern by COVID-19 MRNA Vaccines in Immunocompromised Patients. Jama Oncol. 2022, 8, e220446. [Google Scholar] [CrossRef]
  49. Fendler, A.; Shepherd, S.T.C.; Au, L.; Wilkinson, K.A.; Wu, M.; Byrne, F.; Cerrone, M.; Schmitt, A.M.; Joharatnam-Hogan, N.; Shum, B.; 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, 1305–1320. [Google Scholar] [CrossRef]
  50. Monin, L.; Laing, A.G.; Muñoz-Ruiz, M.; McKenzie, D.R.; del Molino del Barrio, I.; Alaguthurai, T.; Domingo-Vila, C.; Hayday, T.S.; Graham, C.; Seow, J.; 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, 765–778. [Google Scholar] [CrossRef]
  51. Lee, M.; Quinn, R.; Pradhan, K.; Fedorov, K.; Levitz, D.; Fromowitz, A.; Thakkar, A.; Shapiro, L.C.; Kabarriti, R.; Ruiz, R.E.; et al. Impact of COVID-19 on Case Fatality Rate of Patients with Cancer during the Omicron Wave. Cancer Cell 2022, 40, 343–345. [Google Scholar] [CrossRef]
  52. Bakouny, Z.; Labaki, C.; Bhalla, S.; Schmidt, A.L.; Steinharter, J.A.; Cocco, J.; Tremblay, D.A.; Awad, M.M.; Kessler, A.; Haddad, R.I.; et al. Oncology Clinical Trial Disruption during the COVID-19 Pandemic: A COVID-19 and Cancer Outcomes Study. Ann. Oncol. 2022, 33, 836–844. [Google Scholar] [CrossRef]
  53. Jazieh, A.R.; Akbulut, H.; Curigliano, G.; Rogado, A.; Alsharm, A.A.; Razis, E.D.; Mula-Hussain, L.; Errihani, H.; Khattak, A.; De Guzman, R.B.; et al. Impact of the COVID-19 Pandemic on Cancer Care: A Global Collaborative Study. Jco Glob. Oncol. 2020, 6, 1428–1438. [Google Scholar] [CrossRef] [PubMed]
  54. Thomas, S.J.; Perez, J.L.; Lockhart, S.P.; Hariharan, S.; Kitchin, N.; Bailey, R.; Liau, K.; Lagkadinou, E.; Türeci, Ö.; Şahin, U.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  55. Richards, M.; Anderson, M.; Carter, P.; Ebert, B.L.; Mossialos, E. The Impact of the COVID-19 Pandemic on Cancer Care. Nat. Cancer 2020, 1, 565–567. [Google Scholar] [CrossRef] [PubMed]
  56. Wallis, C.J.D.; Catto, J.W.F.; Finelli, A.; Glaser, A.W.; Gore, J.L.; Loeb, S.; Morgan, T.M.; Morgans, A.K.; Mottet, N.; Neal, R.; et al. The Impact of the COVID-19 Pandemic on Genitourinary Cancer Care: Re-Envisioning the Future. Eur. Urol. 2020, 78, 731–742. [Google Scholar] [CrossRef]
  57. Wang, H.; Elsheikh, M.; Gilmour, K.; Cohen, V.; Sagoo, M.S.; Damato, B.; Anguita, R.; Heimann, H.; Hussain, R.; Cauchi, P.; et al. Impact of COVID-19 Pandemic on Eye Cancer Care in United Kingdom. Br. J. Cancer 2021, 124, 1357–1360. [Google Scholar] [CrossRef] [PubMed]
  58. Marcum, M.; Kurtzweil, N.; Vollmer, C.; Schmid, L.; Vollmer, A.; Kastl, A.; Acker, K.; Gulati, S.; Grover, P.; Herzog, T.J.; et al. COVID-19 Pandemic and Impact on Cancer Clinical Trials: An Academic Medical Center Perspective. Cancer Med. 2020, 9, 6141–6146. [Google Scholar] [CrossRef]
  59. Sessa, C.; Cortes, J.; Conte, P.; Cardoso, F.; Choueiri, T.; Dummer, R.; Lorusso, P.; Ottmann, O.; Ryll, B.; Mok, T.; et al. The Impact of COVID-19 on Cancer Care and Oncology Clinical Research: An Experts’ Perspective. Esmo Open 2022, 7, 100339. [Google Scholar] [CrossRef]
  60. SARS-CoV-2 Omicron Variant in Cancer Patients: An Insight into the Vaccine Booster Debate|Future Oncology. Available online: https://www.futuremedicine.com/doi/10.2217/fon-2022-0024 (accessed on 24 September 2022).
  61. COVID-19 Vaccination and Breakthrough Infections in Patients with Cancer—ScienceDirect. Available online: https://www.sciencedirect.com/science/article/pii/S0923753421048808 (accessed on 24 September 2022).
  62. Ranganathan, P.; Sengar, M.; Chinnaswamy, G.; Agrawal, G.; Arumugham, R.; Bhatt, R.; Bilimagga, R.; Chakrabarti, J.; Chandrasekharan, A.; Chaturvedi, H.K.; et al. Impact of COVID-19 on Cancer Care in India: A Cohort Study. Lancet Oncol. 2021, 22, 970–976. [Google Scholar] [CrossRef]
  63. Ingram, S.A.; Caston, N.E.; Andrews, C.J.; England, R.; Williams, C.; Azuero, A.; Gallagher, K.D.; Angove, R.; Anderson, E.; Balch, A.J.; et al. Hesitancy and Malignancy: Vaccine Hesitancy among Individuals with Cancer. JCO 2021, 39, 148. [Google Scholar] [CrossRef]
  64. Ahmad Malik, J.; Ahmed, S.; Shinde, M.; Almermesh, M.H.S.; Alghamdi, S.; Hussain, A.; Anwar, S. The Impact of COVID-19 On Comorbidities: A Review Of Recent Updates For Combating It. Saudi J. Biol. Sci. 2022, 29, 3586–3599. [Google Scholar] [CrossRef]
  65. Chavda, V.P.; Vora, L.K.; Vihol, D.R. COVAX-19Ⓡ Vaccine: Completely Blocks Virus Transmission to Non-Immune Individuals. Clin. Complementary Med. Pharmacol. 2021, 1, 100004. [Google Scholar] [CrossRef]
  66. Chavda, V.P.; Vora, L.K.; Pandya, A.K.; Patravale, V.B. Intranasal Vaccines for SARS-CoV-2: From Challenges to Potential in COVID-19 Management. Drug Discov. Today 2021, 26, 2619–2636. [Google Scholar] [CrossRef]
  67. Chavda, V.P.; Bezbaruah, R.; Athalye, M.; Parikh, P.K.; Chhipa, A.S.; Patel, S.; Apostolopoulos, V. Replicating Viral Vector-Based Vaccines for COVID-19: Potential Avenue in Vaccination Arena. Viruses 2022, 14, 759. [Google Scholar] [CrossRef]
  68. Chavda, V.P.; Patel, A.B.; Vihol, D.; Vaghasiya, D.D.; Ahmed, K.M.S.B.; Trivedi, K.U.; Dave, D.J. Herbal Remedies, Nutraceuticals, and Dietary Supplements for COVID-19 Management: An Update. Clin. Complementary Med. Pharmacol. 2022, 2, 100021. [Google Scholar] [CrossRef]
  69. Huang, Z.; Chavda, V.P.; Vora, L.K.; Gajjar, N.; Apostolopoulos, V.; Shah, N.; Chen, Z.-S. 2-Deoxy-D-Glucose and Its Derivatives for the COVID-19 Treatment: An Update. Front. Pharmacol. 2022, 13. [Google Scholar] [CrossRef] [PubMed]
  70. Chavda, V.P.; Hanuma Kumar Ghali, E.N.; Yallapu, M.M.; Apostolopoulos, V. Therapeutics to Tackle Omicron Outbreak. Immunotherapy 2022, 14, 833–838. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Connection between cancer and COVID-19. (Modified from [16] under CC BY 4.0 License).
Figure 1. Connection between cancer and COVID-19. (Modified from [16] under CC BY 4.0 License).
Vaccines 10 02072 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chavda, V.P.; Ping, F.-F.; Chen, Z.-S. An Impact of COVID-19 on Cancer Care: An Update. Vaccines 2022, 10, 2072. https://doi.org/10.3390/vaccines10122072

AMA Style

Chavda VP, Ping F-F, Chen Z-S. An Impact of COVID-19 on Cancer Care: An Update. Vaccines. 2022; 10(12):2072. https://doi.org/10.3390/vaccines10122072

Chicago/Turabian Style

Chavda, Vivek P., Feng-Feng Ping, and Zhe-Sheng Chen. 2022. "An Impact of COVID-19 on Cancer Care: An Update" Vaccines 10, no. 12: 2072. https://doi.org/10.3390/vaccines10122072

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop