key: cord-1026904-i85fj1zj authors: Focosi, Daniele; Franchini, Massimo title: Potential use of convalescent plasma for SARS-CoV-2 prophylaxis and treatment in immunocompromised and vulnerable populations date: 2021-05-27 journal: Expert review of vaccines DOI: 10.1080/14760584.2021.1932475 sha: 830401a4d845151a625cc7b999e41d0b214dc4b6 doc_id: 1026904 cord_uid: i85fj1zj INTRODUCTION: : The ongoing SARS-CoV-2 pandemic is a serious threat for the health of immunocompromised patients. Among neutralizing antibody-based therapeutics, convalescent plasma containing polyclonal anti-SARS-CoV-2 immunoglobulins has promising results in both congenital and iatrogenic immunodeficiencies in oncohematological and transplant patients. AREAS COVERED: : This article discusses case reports, case series and controlled studies detailing the efficacy of convalescent plasma in immunocompromised patients. EXPERT OPINION: : Convalescent plasma, when administered at high neutralizing antibody titers, is a safe and effective treatment for frail immunocompromised patients. Genetic monitoring of refractory patients is recommended to intercept intra-host emergence of SARS-CoV-2 variants. The COVID19 pandemic has caused more than 125 million cases and 2.5 million deaths worldwide since January 2020. Few drugs have shown robust evidences of clinical efficacy, including a few neutralizing antibody (nAb)-based therapeutics. COVID19 convalescent plasma (CCP), despite poor definition [1] and many controversial trials (summarized in Figure 1 ), is one of the few SARS-CoV-2 therapeutics under massive investigation [2] , having shown clinical benefit when used early (within 72 hours since onset of symptoms) and with high titers of neutralizing antibodies (nAb) [3] [4] [5] . CCP has also proven an extremely safe therapy with very few thromboembolic events [6] . Similarly, monoclonal antibodies (mAb) have been proven effective when administered early in the disease course and in seronegative recipients [7] , but their cost and availability remain severe hurdles. Clinical trials with hyperimmune serum, an industrial derivative of CCP or derived from immunized animals, are still ongoing [8] , but to date no experience has been reported in immunocompromised patients. The absolute requirement for early treatment in frail immunocompromised patients is still questioned, leaving hopes for benefit also in late usages [9] : accordingly, many case reports and series have documented success of CCP in late COVID19 stages in such patients. Long-term benefits still need to be assessed since immunocompromised patients are at risk for reinfection. Similarly, there is a lack of long-term follow-up studies assessing the outcome of CCP therapy on the underlying malignancy or immune deficiency. Preliminary evidences suggest that commercial intravenous immunoglobulin (IVIg) formulations manufactured from plasma donations collected after the beginning of the pandemic could have high titers of anti-SARS-CoV-2 nAb [10] , but no clinical trial has been initiated yet. Frail immunosuppressed patients are prone to more severe COVID19, given the inability to control SARS-CoV-2 infection. The risk profile varies according to the type of immune deficiency (ranging from humoral to cellular to combined T-and B-) and depth (e.g. hypo-vs. agammaglobulinemia). Immunocompromised patients are likely to mount partial or no protective immune response after vaccination, making the availability of effective therapeutics mandatory for such cohort. Unfortunately, these patients have not been represented in large CCP randomized controlled trials (RCT) reported to date [11] , and evidences of efficacy mostly stem from small-scale phase II trials (for oncohematological and transplant patients) or case reports (for rare congenital immune deficiencies). In this narrative review we analyze evidences supporting the efficacy of CCP for post-exposure prophylaxis and early treatment in these frail cohorts. population also affect patients with PID, including more younger patients [12] . Mortality rate was 9% which was similar to the global data from the general population and it was mainly among patients with other comorbidities like heart and kidney diseases [12] . The severity of PID inversely correlate with the severity of COVID-19 [13] . Stricter infection control measures (such as social isolation) compared to the general population likely lead to lower SARS-CoV-2 infection rates in PID patients [12] [13] [14] . Compared to the aggressive clinical course seen in patients with common variable immunodeficiency (CVID), patients with agammaglobulinemia (either Bruton's X-linked (XLA) or autosomal recessive (ARA)) had very mild COVID19 courses [12, 15, 16] . This suggests that T-cell immunity (only moderately affected in XLA and ARA) could partly compensate the lack of neutralizing antibodies. Overall, COVID19 has been reported to date in 13 Bruton's XLA [12, [15] [16] [17] (including one possible reinfection [18] ), 1 ARA [16] , and 11 CVID patients [15, 19, 20] , and 1 with X-SCID [21] . 2 cases have been reported in Good syndrome (thymoma with immunodeficiency) [19, 22] , which is currently classified a phenocopy of PID. Table 1 reports the features of the ones of them who were treated with CCP: of interest, none of them died despite most of them being treated later during the course of the disease. Unfortunately, no data on nAb content in units of in patients were available. Underlying prophylaxis with intravenous immunoglobulins (likely derived from prepandemic donation batches) in patients with hypo/agammaglobulinemias was not sufficient to prevent COVID19, also excluding any beneficial impact from cross-reactive antibodies against seasonal, endemic related coronaviruses. Chronic enterovirus infection is well known in patients with XLA [29] , so it is not unexpected that SARS-CoV-2 can lead to chronic infections in congenital agammaglobulinemias. Immunocompromised patients with hematological cancers have a COVID19 mortality rate as high as 60% [30] [31] [32] . Patients with hematologic malignancies may have immune deficiencies from patient-related (i.e. age), disease-related, and treatment-related (i.e. chemo-immunotherapies) factors. Table 2 summarized the main evidences to date. In a single center cohort of patients with chronic lymphocytic leukemia and symptomatic COVID-19, 7 of 21 (33%) did not develop detectable anti-SARS-CoV-2 antibodies, markedly lower than the 100% seroconversion rate observed in a non-cancer population [33, 34] . Immunocompromised patients suffering from oncohematological cancers, due to their inability to mount an appropriate humoral immune response to SARS-CoV-2, represent the ideal candidate for passive immunotherapy by means of CCP transfusion [35] . There is increasing interest toward the CCP use in patients with hematologic malignancies and several investigators have explored this therapeutic possibility [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] . Details about disease severity, timing of CCP administration, number of doses, pre-and post-treatment nAb titers were unfortunately not available for the vast majority of patients. In a recent case series published by Tremblay and colleagues [54] , the authors identified 24 patients with cancer, 14 of whom with a hematological malignancy (5 non-Hodgkin lymphoma, 1 Hodgkin lymphoma, 4 multiple myeloma, 2 acute lymphoblastic leukemia, 1 myelofibrosis, 1 chronic lymphocytic leukemia), treated with hightiter (≥1:320) CCP within an expanded access protocol. Most patients (62.5%) were on anti-cancer therapy at the time of COVID-19 infection. The overall mortality rate was 41.7% (10/24). Non-intubated patients had favorable outcomes (death rate: 28.5%, 6/21), suggesting a potential clinical benefit of CCP in less advanced stages of COVID-19. In addition, a significant decrease of inflammatory markers (i.e. C-reactive protein, CRP) was observed after 3 days of CCP treatment. Transfusion reactions were uncommon and mild, occurring only in three patients. Of the 14 patients with hematologic malignancies, 8 patients (57.1%) were discharged, 1 (7.1%) was still hospitalized and mechanically ventilated and 5 (35.7%) expired. Hueso and colleagues [37] reported a series of 17 consecutive patients, of whom 15 had hematological malignancies (13 non-Hodgkin lymphoma, 1 chronic lymphocytic leukemia and 1 Waldenstrom macroglobulinemia) with profound B-cell lymphopenia due to anti-CD20 monoclonal antibody therapy and prolonged COVID-19 symptoms, negative SARS-CoV-2 IgG/IgM serology, and positive viral RNA-emia who were treated with 4 units of CCP. No serious adverse effects were observed during or after CCP therapy. Within 48 hours of transfusion, all but 1 patient experienced an improvement of clinical symptoms, including reduced oxygen requirements, which correlated strongly with the viral clearance documented in all the 9 patients evaluated. The hyper-inflammatory status faded within a week. Only 1 patient who needed mechanical ventilation for severe COVID-19 disease died of bacterial pneumonia. The authors concluded that CCP appeared to be a promising therapy for COVID-19 treatment in B-cell depleted patients unable to mount a specific humoral response against SARS-CoV-2. Interestingly, Betrains and colleagues [48] analyzed 5 patients with COVID-19 and B-cell lymphoma treated with anti-CD20 therapy and demonstrated that B-cell depletion was associated with decreased neutralized antibody formation, reduced viral clearance and protracted clinical manifestations of SARS-CoV-2 infection. Treatment with CCP was accompanied by an increase in neutralizing antibody titers in all patients and by a clinical response in all but one patient. The authors concluded that patients with B-celldepleted lymphomas with protracted SARS-CoV-2 infection are the ideal candidates for passive immunotherapy by CCP. Other case reports confirmed this initial finding in similar groups of non-Hodgkin lymphoma patients [45, 47, 49] . In a retrospective analysis, Jeyaraman et al [53] identified 33 patients with hematologic malignancies (18 non-Hodgkin lymphoma, 4 acute leukemia, 7 multiple myeloma, 2 myelodysplastic syndrome, 2 chronic myeloid leukemia) treated for severe COVID-19 with CCP, in the majority of cases within 7 days of COVID-19 diagnosis. The majority of patients were on active chemotherapy (72.7%, 24/33) at the time of CCP infusion. The overall mortality rate in the cohort was 45.5% (15/33) and did not differ between early versus late CCP therapy. The largest experience on this issue is that pre-published by Thompson and colleagues on behalf of the COVID-19 and Cancer Consortium [52] . In this retrospective study, 143 patients with various hematologic malignancies (lymphoid neoplasms 123 and myeloid neoplasms 21) and COVID-19, [50] where the authors reported their own experience on CCP therapy in 7 patients with COVID-19 previously treated with chemo-immunotherapy due to oncohematological disorders (4 non-Hodgkin lymphoma, 1 primary myelofibrosis, 1 chronic lymphocytic leukemia, 1 acute myeloid leukemia). CCP treatment was well tolerated and, in all cases, resulted in a clinical benefit in term respiratory symptoms with less intensive oxygen requirements. Viral clearance was detected by nasopharyngeal swabs in 5 out of 7 patients treated. The authors concluded that CCP can be a safe and effective therapeutic option for oncohematological patients with COVID-19 and immunodeficiency due to previous chemo-immunotherapy. In a recent pre-published review, Senefeld and colleagues [56] identified 54 patients with hematological malignancies, including lymphoma, leukemia, multiple myeloma and myelodysplastic syndrome, transfused with CCP in 18 peer-reviewed reports. A majority of patients recovered following CP transfusion, with many demonstrating rapid clinical improvements shortly after transfusion. Notably, a patient with persistent (> 100 days) COVID-19 and with lymphoma-associated B-cell immunodeficiency demonstrated rapid reductions in fever, oxygen requirements and lung infiltrates immediately after two CCP transfusions separated by approximately 90 days [46] . Recipients of solid organ transplants receiving immunosuppressive medications are at increased risk of severe COVID-19. Only a few case reports or case series have assessed the role of CCP in solid organ transplant patients with COVID-19 [59] [60] [61] [62] . A clinical beneficial effect associated with passive immunotherapy was even seen in the extreme case of a liver transplant recipient with a life-threatening COVID-19: the patient rapidly recovered following a CCP transfusion received during a 17 day medically induced coma due to COVID-19 complications [63]. Fung and colleagues [39] reported 3 transplant patients (2 kidney transplant recipients and 1 lung transplant recipient) treated with CCP through an Expanded Access Program. All patients clinically improved after CCP administration without transfusion reactions. A recent systematic review and meta-analysis by Raja and colleagues [64] identified 215 studies including 2772 transplanted patients (1500 kidney, 505 liver, 141 heart, 97 lung, 1 face and 43 unidentified combined transplants). Although CCP was utilized only in a minority of patients (33 patients enrolled in 13 studies), the mortality rate in this subgroup seemed to be lower than that recorded in the entire population of patients (12.9% versus 18.6%, respectively). In their recent prepublished review, Senefeld and colleagues [56] identified 9 articles reporting 29 COVID-19 patients receiving immunosuppressive therapies for previous solid organ transplants and transfused with hyperimmune plasma. In most case CCP administration was accompanied by improved clinical symptomatology and oxygen requirements and reduction in hospital stay. Despite the scarcity of the literature data and the potential bias of reporting (cases with positive outcomes preferentially reported with respect to those with negative outcomes), the currently reported cases suggest a potential beneficial effect of CP also in this particularly complex category of fragile patients. concern: efficacy and risks of accelerated viral evolution. Spike mutations are rare after immunosuppressive treatment without anti-Spike treatment [65] . Nevertheless Bazykin et al reported emergence of Y453F and Δ69-70HV mutations ('the ΔF combination') (together with S50L, Δ141-144, T470N, and D737G) in a 47-year-old female with diffuse large B cell lymphoma treated with rituximab plus chemotherapy (R-ICE regimen) [66] . Borges et al reported another DLBCL patients with persistent infection for 6 months who developed four mutations (V3G, S50L, N87S and A222V) and two deletions (Δ18-30 and Δ141-144) in Spike [67] . Finally, Truong et al reported the emergence of escape mutations in 2 more patients with acute lymphoblastic leukemia who were persistently positive for SARS-CoV-2 for up to 162 days [68] . Several lines of evidence support the hypothesis that widespread deployment of antibody-based therapeutics could drive Spike immune escape. In vitro evidences include the emergence of mutations during SARS-CoV-2 culture with convalescent plasma. Continuous passaging of SARS-CoV-2 in the presence of a CCP unit with nAb titer >1:10 4 led to ΔF140 at day 45, followed by E484K at days 73 and an insertion in the NTD: these accumulating mutations led to complete lack of neutralization [69] . Accordingly, K417N, E484K, and N501Y mutations. were selected when pseudotyped SARS-CoV-2 was cultured in the presence of the vaccine elicited mAbs [70] . Although within host SARS-CoV-2 mutation accumulation is typically very low [71] , faster rates have been found in longitudinal studies of immunodeficient patients with persistent SARS-CoV-2 infections for up to several months. In particular, this has happened in case reports after treatment with CCP: the phenomenon does not seem very common or very fast, since none out of eight onco-hematological patients (recipients of hematopoietic stem-cell transplants or chimeric antigen receptor T lymphocytes) treated with CCP who remained SARS-CoV-2 positive for 2 months showed significant mutations compared to wild-type strain [65] . Nevertheless, Avanzato et al reported within-host genomic evolution in a patient affected by chronic lymphocytic leukemia and iatrogenic hypogammaglobulinemia treated with CCP and shedding infectious SARS-CoV-2 for 70 days, and subgenomic RNA for 105 days [36] . Similarly, Kemp et al reported an immune suppressed individual who showed little evolutionary change in the first 65 days while on remdesivir, but who developed D796H and ΔH69/ΔV70 mutations twice after 2 unsuccessful courses of CCP. In vitro, such mutant showed similar infectivity to wild type strain but resistance to many CCP donors [72] . Finally, Truong et al reported the emergence of 7 major and 3 minor allele variants (including ∆141-143, ∆145, ∆141-144, ∆211-212, N440K, V483A, and E484Q) in a patient with acute lymphoblastic leukemia who was treated with weekly CCP and tested persistently positive for SARS-CoV-2 until day 144 [68] . Such serial monitoring of immunocompromised patients receiving CCP with next-generation sequencing is extremely expensive and time-consuming. Despite lack of details from these sporadic case reports, we cannot exclude that treatment with subneutralizing antibody levels could have facilitated accelerated viral evolution. In this regard, the introduction of a weight-and titer-adjusted loading dose of CCP seems a prudent approach. Patients showing delayed refractoriness to CCP are encouraged to undergo at least SARS-CoV-2 Spike gene sequencing to exclude variants. COVID19 convalescent plasma (CCP) is a promising drug for treatment and post-exposure prophylaxis of COVID-19. Besides the safety of CCP therapy, which has been recognized by almost all previous trials [2] , the most important issue regards its effectiveness. The great majority of the published literature data strengthen the importance of CCP transfusion in COVID-19 patients as close to symptom onset as possible, in order to promptly block SARS-CoV-2 replication and the consequent progression, often irreversible, of COVID-19 pathology [11] . Another key issue of CCP therapy pertains to the quality of CCP produced by transfusion services. Assuming that the antiviral activity of the CCP is mostly linked to the amount of antibodies present, it follows that the more the neutralizing antibodies, the more effective the plasma will be in blocking viral replication [11] . Although the plaque reduction neutralization test (PRNT), which measures the ability of neutralizing antibodies to prevent infection in vitro calculated as a reduction in the formation of plaques, is the current gold standard to assess viral neutralization by CCP, a number of commercial serological high throughput SARS-CoV-2 assay are being tested to replace PRNT for the determination of CCP neutralizing potency [11] . Although some national and international guidelines [73] recommend the use in immunocompromised patients of CCP at titer ≥ 1:320, the exact dose and the timing of CCP administration are yet to be established and need to be assessed by further trials. Finally, another important factor contributing to the CCP clinical effectiveness regards the COVID-19 patients' characteristics. Controlled studies have shown clinical benefit in iatrogenic immunosuppression (oncohematological patients and solid organ transplant recipients). Among patients with hematological malignancies, those suffering from lymphoproliferative B-cell disorders or receiving B-cell depleting therapies are the ones who are likely to benefit most from CCP therapy. Such patients, indeed, are unable to mount an adequate immune response against SARS-CoV-2 and thus may benefit from the passive transfer of anti-SARS-CoV-2 antibodies through CCP transfusion administered during the early phase of viral infection [35] . Many case reports and cases series have also shown clinical benefit in congenital immune deficiencies, particularly those with genetically determined impaired humoral response. For such reasons, the US Food and Drug Administration (FDA) recently revised the Emergency Use Authorization (EUA) of COVID-19 CP authorizing its use at high titer for the treatment of hospitalized COVID-19 patients early in the course of disease and those hospitalized with impaired humoral immunity [9] . Studies testing CCP as post-exposure prophylaxis are still ongoing, and have the potential to lead to better outcomes than when used as treatment. Given the low chances of mounting a protective immune response after vaccination, nAb-based therapeutics remain a potential drug for early treatment of COVID19 in frail immunocompromised patients. High-titer CCP bulks should hence be maintained, even after reaching herd immunity, for the coming years. The level of protection from commercial aspecific IVIg batches manufactured from plasma donations collected after January 2020 remains to be established. In addition, a number of trials are currently exploring the beneficial effect of specific polyclonal anti-SARS-CoV-2 IVIg [8] . It is important to underline, however, that the production of specific immunoglobulins requires significant investments by the manufacturing companies which take years to recover. It is therefore conceivable that, in the face of herd immunity in the Western world by the end of 2021, the interest of pharmaceutical companies in this type of products will be reduced. This paper was not funded. The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose. All authors should have (1) substantially contributed to the conception and design of the review article and interpreting the relevant literature, and (2) been involved in writing the review article or revised it for intellectual content. http://orcid.org/0000-0001-8811-195X Massimo Franchini http://orcid.org/0000-0002-8795-0580 Urgent need to regulate convalescent plasma differently from thawed plasma Convalescent Plasma Therapy for COVID-19: state of the Art •• This review provides historical background Timing of Convalescent plasma administration and 28-day mortality for COVID-19 pneumonia Convalescent Plasma Antibody Levels and the Risk of Death from Covid-19 Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults This trial provides evidence of benefit in nonhospitalized patients having mild symptoms since less than 3 days Safety Update: COVID-19 Convalescent Plasma in 20,000 Hospitalized Patients REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19 The road towards polyclonal anti-SARS-CoV-2 immunoglobulins (hyperimmune serum) for passive immunization in COVID19. Life (Basel) FDA. Recommendations for Investigational COVID-19 Convalescent Plasma Rapidly Increasing SARS-CoV-2 Neutralization by Intravenous Immunoglobulins Produced from Plasma Collected During the 2020 Pandemic 19 convalescent plasma therapy: hit fast, hit hard! Vox Sang Coronavirus disease 2019 in patients with inborn errors of immunity: an international study •• The largest study on CCP in inherited immunedeficiencies Primary Immunodeficiency Diseases in COVID-19 Pandemic: a Predisposing or Protective Factor? Impact of SARS-CoV-2 Pandemic on Patients with Primary Immunodeficiency A possible role for B cells in COVID-19? Lesson from patients with agammaglobulinemia Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover Three patients with X-linked agammaglobulinemia hospitalized for COVID-19 improved with convalescent plasma COVID-19 reinfection in a patient with X-linked agammaglobulinaemia Severe COVID-19 in Patients with B Cell Alymphocytosis and Response to Convalescent Plasma Therapy The clinical observation of a patient with common variable immunodeficiency diagnosed as having coronavirus disease 2019 SARS-CoV-2 infection associated with hepatitis in an infant with X-linked severe combined immunodeficiency Fatal SARS-CoV-2 infection in a male patient with Good's syndrome Rapid recovery of a SARS-CoV-2-infected X-linked agammaglobulinemia patient after infusion of COVID-19 convalescent plasma Patient with X-Linked Agammaglobulinemia and COVID-19 Infection Treated with Remdesivir and Convalescent Plasma Treatment of COVID-19 with convalescent plasma in patients with humoral immunodeficiency -Three consecutive cases and review of the literature Atypical course of COVID-19 in patient with Bruton agammaglobulinemia Bruton's Agammaglobulinemia and COVID-19 Rapid clinical recovery of a SARS-CoV-2 infected common variable immunodeficiency patient following the infusion of COVID-19 convalescent plasma Enteroviruses in X-Linked Agammaglobulinemia: update on Epidemiology and Therapy Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: a retrospective, multicentre, cohort study COVID-19 in persons with haematological cancers Clinical outcome of coronavirus disease 2019 in haemato-oncology patients Anti-SARS-CoV-2 antibody response in patients with chronic lymphocytic leukemia Antibody responses to SARS-CoV-2 in patients with COVID-19 COVID19 neutralizing antibody-based therapies in humoral immune deficiencies: a narrative review Case Study: prolonged Infectious SARS-CoV-2 Shedding from an Asymptomatic Immunocompromised Individual with Convalescent plasma therapy for B-cell-depleted patients with protracted COVID-19 Concurrent Diagnosis of Acute Myeloid Leukemia and COVID-19: a Management Challenge Treatment of immunocompromised COVID-19 patients with convalescent plasma Convalescent Plasma Rescued a Severe COVID-19 Patient with Chronic Myeloid Leukemia Blast Crisis and Myelofibrosis COVID-19 virus reactivation following treatment for B cell acute lymphoblastic leukemia Successful transfer of anti-SARS-CoV-2 immunity using convalescent plasma in an MM patient with hypogammaglobulinemia and COVID-19 Prolonged viral shedding in a lymphoma patient with COVID-19 infection receiving convalescent plasma Successful Use of COVID-19 Convalescent Plasma in a Patient Recently Treated for Follicular Lymphoma Convalescent plasma for persisting COVID-19 following therapeutic lymphocyte depletion: a report of rapid recovery Prolonged SARS-CoV-2 replication in an immunocompromised patient Sustained response after remdesivir and convalescent plasma therapy in a B-cell depleted patient with protracted COVID-19 Convalescent plasma treatment of persistent severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in patients with lymphoma with impaired humoral immunity and lack of neutralising antibodies A 63-Year-Old Woman with a History of Non-Hodgkin Lymphoma with Persistent SARS-CoV-2 Infection Who Was Seronegative and Treated with Convalescent Plasma Convalescent hyperimmune plasma for chemo-immunotherapy induced immunodeficiency in COVID-19 patients with hematological malignancies SARS-CoV-2 convalescent plasma therapy in pediatric patient after hematopoietic stem cell transplantation Convalescent Plasma and Improved Survival in Patients with Hematologic Malignancies and COVID-19 Convalescent plasma therapy for severe Covid-19 in patients with hematological malignancies Convalescent Plasma for the Treatment of Severe COVID-19 Infection in Cancer Patients Convalescent (immune) plasma treatment in a myelodysplastic COVID-19 patient with disseminated tuberculosis Therapeutic use of convalescent plasma in COVID-19 patients with immunodeficiency Treatment of B-cell depleted COVID-19 patients with convalescent plasma and plasma-based products Potential benefit of convalescent plasma transfusions in immunocompromised patients with COVID-19 Convalescent plasma therapy and remdesivir duo successfully salvaged an early liver transplant recipient with severe COVID-19 pneumonia COVID-19 in recent heart transplant recipients: clinicopathologic features and early outcomes Successful recovery from COVID-19 in three kidney transplant recipients who received convalescent plasma therapy Convalescent plasma therapy: helpful treatment of COVID-19 in a kidney transplant recipient presenting with severe clinical manifestations and complex complications NHS Blood and Transplant N. One of the first COVID-19 convalescent plasma recipients supports donor appeal COVID-19 in solid organ transplant recipients: a systematic review and meta-analysis of current literature Shedding of Viable SARS-CoV-2 after Immunosuppressive Therapy for Cancer Emergence of Y453F and Δ69-70HV mutations in a lymphoma patient with long-term COVID-19 Long-term evolution of SARS-CoV-2 in an immunocompromised patient with non-Hodgkin lymphoma Persistent SARS-CoV-2 infection and increasing viral variants in children and young adults with impaired humoral immunity. bioRxiv SARS-CoV-2 escape in vitro from a highly neutralizing COVID-19 convalescent plasma. bioRxiv mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. biorXiv Temporal dynamics of SARS-CoV-2 mutation accumulation within and across infected hosts Neutralising antibodies in Spike mediated SARS-CoV-2 adaptation Operational protocol for donation of anti-COVID-19 convalescent plasma in Italy