Next Article in Journal
Small Tweaks, Major Changes: Post-Translational Modifications That Occur within M2 Macrophages in the Tumor Microenvironment
Next Article in Special Issue
Strong Humoral but Not Cellular Immune Responses against SARS-CoV-2 in Individuals with Oncohematological Disease Who Were Treated with Rituximab before Receiving a Vaccine Booster
Previous Article in Journal
A Novel m7G-Related Gene Signature Predicts the Prognosis of Colon Cancer
Previous Article in Special Issue
Efficacy of COVID-19 Booster Vaccines in Patients with Hematologic Malignancies: Experiences in a Real-World Scenario
 
 
cancers-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Simultaneous Onset of Haematological Malignancy and COVID: An Epicovideha Survey

by
Chiara Cattaneo
1,†,
Jon Salmanton-García
2,3,*,†,
Francesco Marchesi
4,
Shaimaa El-Ashwah
5,
Federico Itri
6,
Barbora Weinbergerová
7,
Maria Gomes Da Silva
8,
Michelina Dargenio
9,
Julio Dávila-Valls
10,
Sonia Martín-Pérez
10,
Francesca Farina
11,
Jaap Van Doesum
12,
Toni Valković
13,14,15,
Caroline Besson
16,
Christian Bjørn Poulsen
17,
Alberto López-García
18,
Pavel Žák
19,
Martin Schönlein
20,
Klára Piukovics
21,
Ozren Jaksic
22,
Alba Cabirta
23,24,
Natasha Ali
25,
Uluhan Sili
26,
Nicola Fracchiolla
27,
Giulia Dragonetti
28,
Tatjana Adžić-Vukičević
29,
Monia Marchetti
30,
Marina Machado
31,
Andreas Glenthøj
32,
Olimpia Finizio
33,
Fatih Demirkan
34,
Ola Blennow
35,
Maria Chiara Tisi
36,
Ali S. Omrani
37,
Milan Navrátil
38,
Zdeněk Ráčil
39,
Jan Novák
40,
Gabriele Magliano
41,
Moraima Jiménez
42,43,
Carolina Garcia-Vidal
44,
Nurettin Erben
45,
Maria Ilaria Del Principe
46,
Caterina Buquicchio
47,
Rui Bergantim
48,
Josip Batinić
49,50,51,
Murtadha Al-Khabori
52,
Luisa Verga
53,54,
Tomáš Szotkowski
55,
Michail Samarkos
56,
Irati Ormazabal-Vélez
57,
Stef Meers
58,
Johan Maertens
59,
László Imre Pinczés
60,
Martin Hoenigl
61,62,63,
Ľuboš Drgoňa
64,
Annarosa Cuccaro
65,
Yavuz M. Bilgin
66,
Avinash Aujayeb
67,
Laman Rahimli
2,3,
Stefanie Gräfe
2,3,68,
Mariarita Sciumè
27,
Miloš Mladenović
29,69,
Gökçe Melis Çolak
26,
Maria Vittoria Sacchi
30,
Anna Nordlander
35,
Caroline Berg Venemyr
32,
Michaela Hanáková
39,
Nicole García-Poutón
43,
Ziad Emarah
5,
Giovanni Paolo Maria Zambrotta
52,53,
Raquel Nunes Rodrigues
8,
Raul Cordoba
18,
Gustavo-Adolfo Méndez
70,
Monika M. Biernat
71,
Oliver A. Cornely
2,3,72,73,‡ and
Livio Pagano
28,74,‡
add Show full author list remove Hide full author list
1
Hematology Unit, ASST-Spedali Civili, 25123 Brescia, Italy
2
University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
3
University of Cologne, Faculty of Medicine, University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), 50931 Cologne, Germany
4
Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
5
Oncology Center, Mansoura University, Mansoura 35516, Egypt
6
San Luigi Gonzaga Hospital, 10043 Orbassano, Italy
7
Masaryk University and University Hospital Brno—Department of Internal Medicine, Hematology and Oncology, 62500 Brno, Czech Republic
8
Portuguese Institute of Oncology, 1099-023 Lisbon, Portugal
9
Hematology and Stem Cell Transplan Unit, Vito Fazzi, 73100 Lecce, Italy
10
Hospital Nuestra Señora de Sonsoles, 05004 Ávila, Spain
11
IRCCS Ospedale San Raffaele, 20132 Milan, Italy
12
University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
13
University Hospital Centre Rijeka, 51000 Rijeka, Croatia
14
Croatian Cooperative Group for Hematological Diseases (CROHEM), 10000 Zagreb, Croatia
15
Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
16
Centre Hospitalier de Versailles, Le Chesnay, Université Paris-Saclay, UVSQ, Inserm, Équipe “Exposome et Hérédité”, CESP, 94800 Villejuif, France
17
Zealand University Hospital, 4000 Roskilde, Denmark
18
Fundacion Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, 28040 Madrid, Spain
19
University Hospital Hradec Králové, 50005 Hradec Králové, Czech Republic
20
Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
21
Department of Internal Medicine, South Division, Faculty of Medicine University of Szeged, 6720 Szeged, Hungary
22
University Hospital Dubrava, 10000 Zagreb, Croatia
23
Department of Hematology, Vall d’Hebron Hospital Universitari, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, 08193 Barcelona, Spain
24
Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
25
Aga Khan University, 74000 Karachi, Pakistan
26
Department of Infectious Diseases and Clinical Microbiology, School of Medicine, Marmara University, 34854 Istanbul, Turkey
27
Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
28
Hematology Unit, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, 00168 Rome, Italy
29
COVID Hospital “Batajnica”, 11000 Belgrade, Serbia
30
Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
31
Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain
32
Department of Hematology, Copenhagen University Hospital—Rigshospitalet, DK-2100 Copenhagen, Denmark
33
UOC Hematology, AORN Cardarelli, 80131 Naples, Italy
34
Dokuz Eylul University, Division of Hematology, 35340 Izmir, Turkey
35
Department of Infectious Diseases, Karolinska University Hospital, 17176 Stockholm, Sweden
36
Ospedale San Bortolo, 36100 Vicenza, Italy
37
Communicable Disease Center, Hamad Medical Corporation, Doha 3050, Qatar
38
University Hospital Ostrava, 70852 Ostrava, Czech Republic
39
Institute of Hematology and Blood Transfusion, 12800 Prague, Czech Republic
40
Department of Hematology, University Hospital Kralovske Vinohrady and Third Faculty of Medicine, Charles University, 12808 Prague, Czech Republic
41
ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
42
Department of Hematology, University Hospital Vall d’Hebron, Experimental Hematology, 08035 Barcelona, Spain
43
Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
44
Department of Infectious Diseases, Hospital Clinic de Barcelona, University of Barcelona, IDIBAPS, 08035 Barcelona, Spain
45
Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
46
Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
47
Ematologia Con Trapianto, Ospedale Dimiccoli Barletta, 70051 Barletta, Italy
48
Centro Hospitalar e Universitário São João, 4200-319 Porto, Portugal
49
University Hospital Centre Zagreb, 10000 Zagreb, Croatia
50
Croatian Cooperative Group for Hematological Diseases (KROHEM), 10000 Zagreb, Croatia
51
School of Medicine University of Zagreb, 10000 Zagreb, Croatia
52
Sultan Qaboos University Hospital, Muscat 123, Oman
53
Azienda Ospedaliera San Gerardo—Monza, 20900 Monza, Italy
54
Università Milano-Bicocca, 20126 Milan, Italy
55
University Hospital Olomouc, 77900 Olomouc, Czech Republic
56
Laikon Hospital, 11527 Athens, Greece
57
Hospital Universitario de Navarra, 31008 Iruña-Pamplona, Spain
58
AZ KLINA, 2930 Brasschaat, Belgium
59
KU Leuven, 3000 Leuven, Belgium
60
Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
61
Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
62
Clinical and Translational Fungal-Working Group, University of California San Diego, La Jolla, CA 92093, USA
63
Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria
64
Department of Oncohematology, Comenius University and National Cancer Institute, 81499 Bratislava, Slovakia
65
Hematology Unit, Center for Translational Medicine, Azienda USL Toscana NordOvest, 55100 Livorno, Italy
66
Department of Internal Medicine, ADRZ, 4462 RA Goes, The Netherlands
67
Northumbria Healthcare, Newcastle NE29 8NH, UK
68
Universitätsklinikum Hamburg Eppendorf, 20251 Hamburg, Germany
69
Clinic for Orthopedic Surgery and Traumatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
70
Hospital Escuela de Agudos Dr. Ramón Madariaga, Posadas 3736, Argentina
71
Department of Haematology, Blood Neoplasms, and Bone Marrow Transplantation, Wroclaw Medical University, 50-425 Wroclaw, Poland
72
University of Cologne, Faculty of Medicine, University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), 50931 Cologne, Germany
73
German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
74
Hematology Unit, Università Cattolica del Sacro Cuore, 86100 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cancers 2022, 14(22), 5530; https://doi.org/10.3390/cancers14225530
Submission received: 11 October 2022 / Revised: 27 October 2022 / Accepted: 4 November 2022 / Published: 10 November 2022
(This article belongs to the Special Issue COVID-19 Infection and Hematological Malignancies)

Abstract

:

Simple Summary

Patients with simultaneous diagnosis of haematological malignancies (HM) and COVID-19 are an even greater challenge for hematologists. To better clarify their outcome, we describe the clinical features and outcome of a cohort of 450 patients with simultaneous diagnosis of HM and COVID-19 registered in the EPICOVIDEHA registry between March 2020 to February 2022. Overall, 343 (76.2%) patients received treatment for HM, and an overall response rate was observed in 140 (40.8%) patients after the first line of treatment. Thirty-day mortality was significantly higher in patients not receiving HM treatment (42.1%) than in those receiving treatment (27.4%, p = 0.004). Statistical analysis showed that, together with age, severe/critical COVID-19, ≥2 comorbidities, lack of HM treatment was an independent risk factors for mortality. These observations suggest the importance of HM treatment in these patients; therefore, it should be delivered as soon as possible for patients requiring immediate therapy.

Abstract

Background: The outcome of patients with simultaneous diagnosis of haematological malignancies (HM) and COVID-19 is unknown and there are no specific treatment guidelines. Methods: We describe the clinical features and outcome of a cohort of 450 patients with simultaneous diagnosis of HM and COVID-19 registered in the EPICOVIDEHA registry between March 2020 to February 2022. Results: Acute leukaemia and lymphoma were the most frequent HM (35.8% and 35.1%, respectively). Overall, 343 (76.2%) patients received treatment for HM, which was delayed for longer than one month since diagnosis in 57 (16.6%). An overall response rate was observed in 140 (40.8%) patients after the first line of treatment. After a median follow-up of 35 days, overall mortality was 177/450 (39.3%); 30-day mortality was significantly higher in patients not receiving HM treatment (42.1%) than in those receiving treatment (27.4%, p = 0.004), either before and/or after COVID-19, or compared to patients receiving HM treatment at least after COVID-19 (15.2%, p < 0.001). Age, severe/critical COVID-19, ≥2 comorbidities, and lack of HM treatment were independent risk factors for mortality, whereas a lymphocyte count >500/mcl at COVID-19 onset was protective. Conclusions: HM treatment should be delivered as soon as possible for patients with simultaneous diagnosis of COVID-19 and HM requiring immediate therapy.

1. Introduction

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was declared a global pandemic by the World Health Organization (WHO) in March 2020. Since then, many reports demonstrated high morbidity and mortality in haematological patients with coronavirus disease 2019 (COVID-19), partly driven by uncontrolled haematological malignancy (HM), which can affect the severity of COVID-19 [1,2,3].
However, to date, few data are available about the impact of starting early versus delayed specific HM treatment as well as the outcome of patients with simultaneous onset of HM and COVID-19. Some recommendations from expert panels have raised concern regarding the management of specific HM, including acute leukaemia (AL), multiple myeloma (MM) and hairy cell leukaemia (HCL), during the pandemic [4,5,6,7,8]. In these recommendations, few suggestions apply to patients with simultaneous HM and COVID-19 diagnosis, and the advice is usually to delay, when possible, the start of HM therapy. Indeed, the severity of the COVID-19 clinical picture might make any HM specific therapeutic approach impossible, but some cases of simultaneous diagnosis of COVID-19 and AL requiring immediate treatment have been reported, suggesting that the HM therapeutic approach may be possible even with an active infection [9,10]. Other researchers have indicated that an intensity-reduced approach may be helpful, particularly in the management of specific HM, such as acute myeloid leukaemia (AML) [11].
Specific data on a large cohort of patients with simultaneous onset of HM and COVID-19 with long-term follow-up are still lacking, and there are no evidence-based algorithms to guide clinicians in the selection of the best therapeutic approach and timing, particularly for patients requiring urgent antineoplastic treatment.
Hereby, we aimed to assess the overall survival (OS) of patients with simultaneous onset of HM and COVID-19 as primary endpoint of the study. In addition, we intended to determine: (1) the HM response rate of patients with simultaneous onset of HM and COVID-19; (2) the rate of patients receiving specific treatment for HM during the acute phase of COVID-19; (3) the prognostic factors in patients with simultaneous onset of HM and COVID-19; (4) the time interval between HM diagnosis and initiation of therapy; (5) the safety of specific haematological treatments during COVID-19.

2. Materials and Methods

We have conducted an observational multicentre analysis of patients with HM who developed COVID-19 between March 2020 and February 2022. Data were collected from the EPICOVIDEHA registry. EPICOVIDEHA (www.clinicaltrials.gov; NCT04733729) is an international open web-based registry for patients with HM infected with SARS-CoV-2. This registry was centrally approved by the local ethics committee of the Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore of Rome, Italy (Study ID: 3226). Additionally, if applicable, the respective local ethics committee of each participating institution might have approved the EPICOVIDEHA. EPICOVIDEHA’s methods have been described elsewhere [12]. The electronic case report form (eCRF) is accessible online at www.clinicalsurveys.net (EFS Summer 2021, TIVIAN, Cologne, Germany, Accessed on 23 February 2022). Each documented patient was reviewed and validated by experts in infectious diseases and haematology. Inclusion criteria were described as follows: (a) new diagnosis of HM within 33 days before or 3 days after SARS-CoV-2 infection diagnosis, (b) adult patients ≥ 18 years old, and (c) laboratory-based diagnosis of SARS-CoV-2 infection.
Data on patient baseline conditions pre-COVID-19 (i.e., age, sex, factors predisposing for COVID-19, COVID-19 vaccination status), HM clinical management (i.e., type of treatment received and any delay), COVID-19 severity, and outcome (i.e., mortality, attributable mortality [assessed by the medical team in charge of the patient], last day of follow-up) were collected. COVID-19 severity was graded according to international standards as previously described (asymptomatic: no clinical signs or symptoms; mild: non-pneumonia and mild pneumonia; severe: dyspnoea, respiratory frequency ≥ 30 breaths per min, SpO2 ≤ 93%, PaO2/FiO2 < 300, or lung infiltrates > 50%); critical: patients admitted to intensive care for respiratory failure, septic shock, or multiple organ dysfunction or failure) [1] HM treatment was considered delayed if delivered more than one month after the HM diagnosis. Overall response rate for HM included complete remission and partial remission [13]. Neutropenia and lymphopenia were reported based on grade 3 and 4 values according to Common Terminology Criteria for Adverse Events (CTCAE) v6.0 [14]. The observation period was split into four intervals (1st: March 2020–September 2020; 2nd: October 2020–February 2021; 3rd: March 2021–November 2021; 4th: December 2021–February 2022), according to pandemic waves and to vaccine availability.
No a priori sample size calculation was performed for this analysis. Categorical variables are described using frequencies and percentages, whereas continuous variables using median, interquartile range (IQR) and absolute range. A Cox regression hazard model was designed and run with variables suspected to play a role in the mortality of patients with COVID-19, as previously described [11]. A multivariable Cox regression model was calculated with the Wald backward method, including only the variables statistically significant in the univariable model. Mortality was analysed using Kaplan–Meier survival plots. Log-rank test was used to compare the survival probability of the patients included in the different plots. A p-value ≤ 0.05 was found statistically significant. SPSSv27.0 was employed for statistical analyses (SPSS, IBM Corp., Chicago, IL, USA).

3. Results

3.1. Characteristics of Patients

From March 2020 to February 2022, 450 patients with simultaneous onset of HM and COVID-19 have been reported in the EPICOVIDEHA registry. Of these, 118 (26.2%) patients were diagnosed with COVID-19 between March and September 2020, 167 (37.1%) between October 2020 and February 2021, 71 (15.8%) between March and November 2021, and 94 (20.9%) between December 2021 and February 2022.
Patient characteristics are summarized in Table 1. Males were predominant (n = 264, 58.7%), with a median age of 65 years (IQR: 53 to 68). At least one comorbidity was reported in 140 (62.7%) patients, and two in 142 (31.6%). AL, particularly AML, were the most common HM (AL n = 161, 35.8%; AML n = 129, 28.7%), together with non-Hodgkin Lymphoma (NHL), mainly aggressive (NHL n = 142, 31.6%; aggressive NHL n = 91, 20.2%). The median time between HM and COVID-19 diagnosis was 11 days (IQR: −22 to −2) before COVID-19. At COVID-19 diagnosis, neutropenia was present in 97 (21.5%) patients (grade 4 in n = 69, 15.5% with less than 500 neutrophils/mm3), and lymphopenia in 101 (22.4%) patients. Secondary infections were reported in 63 (14%) of patients; of these, 6 (9.5%) were invasive possible or probable mould diseases. One fifth of the patients (n = 89, 19.8%) received at least one vaccine dose before COVID-19 diagnosis, with 29 (6.4%) being fully vaccinated; they were recorded all in the 3rd and 4th intervals. In 70 cases patients were vaccinated with mRNA based vaccines. COVID-19 was severe or critical in 286 (63.6%) patients. Most of the patients were admitted to hospital (n = 380, 84.4%); a specific treatment for COVID-19 was delivered in 173 (38.4%) patients, as reported in Table 1. In Table S1 COVID-19 directed antivirals and monoclonal antibodies are reported.

3.2. HM Treatment

Overall, at least 343 (76.2%) patients with simultaneous COVID-19 and HM received treatment for HM, either before and/or after COVID-19 diagnosis. HM treatment was delivered at least before COVID-19 diagnosis in 227/343 (66.2%) patients, with a median time between HM diagnosis and start of HM treatment of 7 days (IQR: 3 to 19). HM treatment was started only after COVID-19 in 116 (33.8%) patients. Only 41 (5.6%) patients were treated from day 32 of COVID-19 onwards. Forty-five (10.0%) patients were treated only with palliative care, before or after COVID-19. One-hundred and seven (23.7%) patients did not receive any treatment until the last day of follow-up. In seven (1.5%) cases no information about HM treatment was provided.
HM treatment was delayed beyond one month since HM diagnosis in 57 (17.2%) patients. Treatment delay was rarer for AL (AML: 9/103, 8.7%, acute lymphoblastic leukaemia, ALL: 2/27, 9.1%) and MM (5/41, 12.2%) and more frequent in NHL (27/112, 21.7%), both aggressive (21.9%) and indolent (20.8%), myelodysplastic/myeloproliferative syndromes (MDS/MPS) (5/22, 22.7%), chronic lymphoproliferative disorders (CLD) (4/11, 36.4%) and Hodgkin lymphoma (HL) (7/14, 50%). In Supplementary Table S2 all the treatments for HM delivered before and after COVID-19 diagnosis are detailed.
Table 2 summarizes the clinical characteristics of patients by HM treatment. Patients receiving HM treatment (either before or after COVID-19) were more often female. AML or lymphoma patients were more frequently treated than those with CLD or MDS/MPS. The number of comorbidities and COVID-19 severity, as well as vaccination status and specific treatment for COVID-19 did not differ significantly in treated and untreated patients. The largest number of HM treated patients was observed in the second period (October 2020–February 2021). Thirty-day mortality was significantly lower in treated patients for HM as compared to those receiving no HM treatment.

3.3. Patient Outcome

Among the 343 treated patients, HM treatment overall response rate was 40.8% (140/343 patients) after the first line of treatment; 100 (29.2%) patients had complete remission. Chemotherapy-induced neutropenia occurred in 155 (45.2%) patients and secondary infections were reported in 85 (24.8%).
After a median follow-up of 35 days (IQR: 12-168) since COVID-19 diagnosis, 177/450 (39.3%) had died. Thirty-day mortality was 139/450 (30.9%) for the whole cohort. It was higher in AML (57/129, 44.2%) and in MM patients (21/55, 38.2%) and lower in lymphoma (39/156, 25%), in MDS/MPS (9/39, 23.1%), in ALL (6/32, 18.7%) and in CLD (7/39, 17.9%) patients (overall survival probability reported in Figure 1a,b). Thirty-day mortality was similar in indolent and aggressive NHL (respectively: 12/42, 28.6% and 26/91, 28.6%), regardless of different proportions of HM treatment received (24/42, 57.1% and 81/91, 90.1%, respectively). In 151 (85.3%) patients COVID-19 was responsible for death; however, in 78 (44%) patients the main reason for death was COVID-19, in 73 (41.2%) COVID-19 and HM and in 26 (5.8%) HM. Thirty-day mortality was significantly higher in patients not receiving HM treatment (42.1%) than in those receiving treatment (27.4%, p = 0.004), either before and/or after COVID-19, or compared to patients receiving HM treatment at least after COVID-19 (15.2%, p < 0.001). The lowest mortality (12.1%) was observed in patients receiving treatment only after COVID-19. Mortality rates by timing of HM treatment are showed in Table S3.
Overall, patients treated for HM showed a higher survival probability at 30 days follow-up as compared to those untreated (p < 0.001) (Figure 2). Higher survival probability in those who were treated was also found in patients with AML (p < 0.001), lymphoma (p < 0.001), MDS/MPS (p = 0.01), and MM (p = 0.052) (Figure S1).

3.4. Risk Factors for Mortality

Increasing age, AML diagnosis, pre-existing comorbidities, critical COVID-19, and no HM treatment were observed as risk factors for mortality in the univariable analysis, whereas simultaneous HM and COVID-19 diagnosis in the 2nd and 4th period, a lymphocyte count > 500/mcl at COVID-19 onset, administration of at least conventional chemotherapy, administration of at least immunotherapy before COVID-19, HM treatment after COVID-19, and anti-SARS-CoV-2 monoclonal antibody administration were protective. At multivariable analysis, age, report of at least two comorbidities, severe/critical COVID-19, and no treatment were independently associated with an increased mortality, whereas a lymphocyte count >500/mcl at COVID-19 onset was protective. Chemotherapy induced neutropenia during the acute phase of COVID-19 did not show any impact on mortality (Table 3).
Age, critical COVID-19, and no HM treatment were risk factors for mortality at univariable and multivariable analyses for both AML and lymphoma patients. Chemotherapy induced neutropenia during the acute phase of COVID-19 was not a mortality risk factor in AML cases, whereas it was for lymphoma patients; immunotherapy before COVID-19 was protective at univariable analysis for lymphoma patients. Moreover, the diagnosis of COVID-19 in consecutive waves to the initial one was associated with a reduced risk for mortality as compared to the first in lymphoma patients at multivariable analysis, while this was not significant for AML (Tables S4 and S5).

4. Discussion

The COVID-19 pandemic has impacted the algorithms for treating HM. While there is some consensus on when to avoid or delay HM treatment during the pandemic [4], no guidelines are available yet for treatment of patients with simultaneous COVID-19 and HM diagnosis. Here we present results of a dataset of 450 patients with simultaneous diagnosis of HM and COVID-19, derived from the EPICOVIDEHA registry. In this analysis, AL and lymphoma were predominant, and about three quarters of these patients received treatment for HM. An overall response rate was observed in about 40% of patients after first-line treatment, and overall survival was 60.7%. Age, critical COVID-19, ≥2 comorbidities and lack of HM treatment were independent predictors of mortality.
The demographic characteristics of patients were similar to those observed in the overall population of the EPICOVIDEHA registry [1] and in other series [2,3]. Most patients were admitted to hospital, with a very high proportion of AL patients, particularly AML, which could be justified by the clinical complexity of these patients who cannot be managed at home since the onset of HM. Indeed, more than half of patients showed a severe or critical COVID-19, which is the subset more difficult to treat for the haematologists.
Among patients receiving HM treatment, 66.2% initiated their treatment before COVID-19 diagnosis and only 33.8% after. As compared to non-treated patients, treated patients were more often female and younger, which could be explained by a relative lower severity of COVID-19. Treated patients were more often affected by AL and, although to a lesser extent, lymphoma, probably because of the urgent need of treatment for these patients. Severity of COVID-19 did not differ significantly in treated and untreated patients, whereas the degree of severity was less pronounced when considering patients treated at least after COVID-19. Thirty-day mortality was significantly lower in patients treated either before and/or after COVID-19 or only after COVID-19 compared to patients with no treatment at all, suggesting that early HM treatment contributes to a better control of SARS-CoV-2 infection.
Overall, the response rate to HM treatment was relatively low (40.8%), probably also for the proportion of deceased patients during treatment, and after a median follow-up of 35 days 60.7% of patients remained alive. COVID-19 alone or in association with HM was the main cause of death. As expected, AML was the HM with the highest 30-day mortality (44.2%), confirming data already reported in the pre-vaccination era [1,5,15]. Mortality was markedly lower (18.7%) in ALL. Causes of this difference may be found in the usually younger age of ALL patients as compared to AML and in the less intensive induction regimen required for ALL treatment than AML, in addition to the fact that also steroid alone can control the disease, at least for a limited period. A Spanish study [16] reported a mortality of 29% in an ALL cohort including patients on salvage treatment during the first two COVID-19-waves. A lower mortality (11.1%) in ALL has also been reported by Chiaretti et al. [17] in a series of 63 ALL COVID-19 patients in a recent observational study, where only a minority (22.2%) of patients became infected with SARS-CoV-2 at diagnosis.
Considering lymphoproliferative disorders, a wide range of mortality rates has been observed among MM, lymphoma and CLD patients in previous EPICOVIDEHA studies [1], with the worst prognosis for MM. As reported in other series enrolling patients during different phases of HM [2,3], it is interesting to notice that no difference in mortality has been recorded between aggressive and indolent NHL COVID-19 patients neither at HM onset, probably reflecting an intrinsic immune response impairment of NHL patients.
Lack of HM treatment was an independent risk factor for mortality, in addition to well-established risks such as age, COVID-19 severity and presence of comorbidities. This association was quite evident in some HM patients, particularly AML, lymphoma, MDS/MPS and MM; it highlights the crucial role of early HM treatment and, therefore, of disease control, for a favourable outcome. In our series, chemotherapy induced neutropenia during the acute phases of COVID-19 did not affect the outcome of AML patients, as opposed to lymphoma patients, where it did. The role of neutropenia as negative impact on AML patients is still a matter of debate. Although some series report a negative impact of neutropenia [18,19,20,21], in the AML EPICOVIDEHA study [5] neutropenia at onset of COVID-19 was not a risk factor for mortality. Indeed, neutropenia is an intrinsic feature of AML, and it is present in AML patients regardless of treatment received. This is not true for lymphoma patients, where chemotherapy induced neutropenia had a negative impact on survival, confirming that less intensive treatment should be preferred during the acute phase of COVID-19, as also suggested by the recent ECIL recommendations [22]. The incidence of secondary infections was relatively low (14%), and invasive fungal diseases were even rarer. These findings are in line with other reports on ICU patients [23,24] or HM patients [25].
In our cohort, less than ten percent of patients were fully vaccinated against COVID-19. Therefore, our study was not able to appropriately evaluate the impact of vaccination on outcome. We also analysed the outcome according to the pandemic period and we observed a relationship only for lymphoma patients, where any period after the first was independently associated to a better outcome. The mortality risk reduction across pandemic periods could be explained by multiple factors, such as an increased number of asymptomatic or mild infections registered while the pandemic was progressing and maybe also a lower virulence of SARS-CoV-2 infection detected over time [26], while in this study COVID-19 specific treatments did not have a relevant impact. Interestingly, in contrast to some of the largest epidemiologic studies published so far [1,5], no impact of pandemic period was observed for AML and simultaneous COVID-19 diagnosis patients, probably because of the burden of active HM disease.
The study is subject to several limitations. First, the absence of a comparison group of HM patients without COVID-19 makes it difficult to assess the real impact of covid on the prognosis of haematological disease and vice versa. Moreover, the choice of the time frame (−33 days to +3 days from COVID-19) for the simultaneous diagnosis of HM and COVID-19 was chosen based on the enrolment criteria of the protocol, which planned to record only patients who had a history of HM. We decided to not go beyond 33 days before COVID-19 to avoid enrolling patients who have already undergone more than one course of HM treatment. Another limitation of the study is the heterogeneity of the HM population and the different algorithms of HM treatment adopted in different countries during the COVID-19 pandemic, which makes interpretation of the results more difficult. Indeed, the concept of treatment delay is not the same for aggressive and indolent HM diseases. Another limitation is that we do not know exactly how many patients went from curative intent to palliation after COVID-19. Moreover, the inclusion of patients in the registry was potentially influenced by the impact of the pandemic waves, and results may not be applicable to the current omicron wave. Finally, the low percentage of vaccinated cases as well as those receiving COVID-19 treatment with monoclonal antibodies limit the applicability to the current and potentially also future phases of the pandemic.

5. Conclusions

Patients with simultaneous COVID-19 and HM still represent a challenge for haematologists, particularly in elderly patients with AML. Our results outline the importance of HM treatment delivered as soon as possible for patients requiring immediate HM therapy. Indeed, Haematologists may fear initiating treatment that may cause more immunosuppression or lead to other side effects in a patient with active COVID-19; however, the consequences of delayed or suspended treatment may be worse. Any supportive measure should be provided for those patients with chemotherapy-induced neutropenia. Whenever possible, a less-intensive approach should be considered, particularly for lymphoma patients, where chemotherapy-induced neutropenia was found as risk factor for mortality. Future studies should evaluate the impact of large-scale COVID-19 vaccination on outcome of patients with simultaneous HM and COVID-19 diagnosis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14225530/s1, Table S1: COVID-19 directed antivirals; Table S2: HM treatments before and after COVID-19 diagnosis; Table S3: 30-day mortality according to HM treatment start; Table S4: Univariable and multivariable analysis for risk factors for mortality (AML patients); Table S5: Univariable and multivariable analysis for risk factors for mortality (Lymphoma patients); Figure S1: 30-day survival probability of treated vs. untreated patients according to HM.

Author Contributions

C.C., J.S.-G., O.A.C. and L.P. contributed to study design, study supervision, and data interpretation and wrote the paper C.C., J.S.-G. and L.P. did the statistical plan. J.S.-G. performed the analysis and C.C., J.S.-G. and L.P. interpreted the data. C.C., J.S.-G., F.M., S.E.-A., F.I., B.W., M.G.D.S., M.D., J.D.-V., S.M.-P., F.F., J.V.D., T.V., C.B. (Caroline Besson), C.B.P., A.L.-G., P.Ž., M.S. (Martin Schönlein), K.P., O.J., A.C. (Alba Cabirta), N.A., U.S., N.F., G.D., T.A.-V., M.M. (Monia Marchetti), M.M. (Marina Machado), A.G., O.F., F.D., O.B., M.C.T., A.S.O., M.N., Z.R., J.N., G.M., M.J., C.G.-V., N.E., M.I.D.P., C.B. (Caterina Buquicchio), R.B., J.B., M.A.-K., L.V., T.S., M.S. (Michail Samarkos), I.O.-V., S.M., J.M., L.I.P., M.H. (Martin Hoenigl), Ľ.D., A.C. (Annarosa Cuccaro), Y.M.B., A.A., L.R., S.G., M.S. (Mariarita Sciumè), M.M. (Miloš Mladenović), G.M.Ç., M.V.S., A.N., C.B.V., M.H. (Michaela Hanáková), N.G.-P., Z.E., G.P.M.Z., R.N.R., R.C., G.-A.M., M.M.B., O.A.C. and L.P. recruited participants and collected and interpreted data. C.C., J.S.-G., F.M., S.E.-A., F.I., B.W., M.G.D.S., M.D., J.D.-V., S.M.-P., F.F., J.V.D., T.V., C.B. (Caroline Besson), C.B.P., A.L.-G., P.Ž., M.S. (Martin Schönlein), K.P., O.J., A.C. (Alba Cabirta), N.A., U.S., N.F., G.D., T.A.-V., M.M. (Monia Marchetti), M.M. (Marina Machado), A.G., O.F., F.D., O.B., M.C.T., A.S.O., M.N., Z.R., J.N., G.M., M.J., C.G.-V., N.E., M.I.D.P., C.B. (Caterina Buquicchio), R.B., J.B., M.A.-K., L.V., T.S., M.S. (Michail Samarkos), I.O.-V., S.M., J.M., L.I.P., M.H. (Martin Hoenigl), Ľ.D., A.C. (Annarosa Cuccaro), Y.M.B., A.A., L.R., S.G., M.S. (Mariarita Sciumè), M.M. (Miloš Mladenović), G.M.Ç., M.V.S., A.N., C.B.V., M.H. (Michaela Hanáková), N.G.-P., Z.E., G.P.M.Z., R.N.R., R.C., G.-A.M., M.M.B., O.A.C. and L.P. contributed to manuscript writing and review of the manuscript. C.C., J.S.-G., F.M., S.E.-A., F.I., B.W., M.G.D.S., M.D., J.D.-V., S.M.-P., F.F., J.V.D., T.V., C.B. (Caroline Besson), C.B.P., A.L.-G., P.Ž., M.S. (Martin Schönlein), K.P., O.J., A.C. (Alba Cabirta), N.A., U.S., N.F., G.D., T.A.-V., M.M. (Monia Marchetti), M.M. (Marina Machado), A.G., O.F., F.D., O.B., M.C.T., A.S.O., M.N., Z.R., J.N., G.M., M.J., C.G.-V., N.E., M.I.D.P., C.B. (Caterina Buquicchio), R.B., J.B., M.A.-K., L.V., T.S., M.S. (Michail Samarkos), I.O.-V., S.M., J.M., L.I.P., M.H. (Martin Hoenigl), Ľ.D., A.C. (Annarosa Cuccaro), Y.M.B., A.A., L.R., S.G., M.S. (Mariarita Sciumè), M.M. (Miloš Mladenović), G.M.Ç., M.V.S., A.N., C.B.V., M.H. (Michaela Hanáková), N.G.-P., Z.E., G.P.M.Z., R.N.R., R.C., G.-A.M., M.M.B., O.A.C. and L.P. agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. C.C., J.S.-G., F.M., S.E.-A., F.I., B.W., M.G.D.S., M.D., J.D.-V., S.M.-P., F.F., J.V.D., T.V., C.B. (Caroline Besson), C.B.P., A.L.-G., P.Ž., M.S. (Martin Schönlein), K.P., O.J., A.C. (Alba Cabirta), N.A., U.S., N.F., G.D., T.A.-V., M.M. (Monia Marchetti), M.M. (Marina Machado), A.G., O.F., F.D., O.B., M.C.T., A.S.O., M.N., Z.R., J.N., G.M., M.J., C.G.-V., N.E., M.I.D.P., C.B. (Caterina Buquicchio), R.B., J.B., M.A.-K., L.V., T.S., M.S. (Michail Samarkos), I.O.-V., S.M., J.M., L.I.P., M.H. (Martin Hoenigl), Ľ.D., A.C. (Annarosa Cuccaro), Y.M.B., A.A., L.R., S.G., M.S. (Mariarita Sciumè), M.M. (Miloš Mladenović), G.M.Ç., M.V.S., A.N., C.B.V., M.H. (Michaela Hanáková), N.G.-P., Z.E., G.P.M.Z., R.N.R., R.C., G.-A.M., M.M.B., O.A.C. and L.P. have read and agreed to the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

EPICOVIDEHA has received funds from Optics COMMITTM (COVID-19 Unmet Medical Needs and Associated Research Extension) COVID-19 RFP program by GILEAD Science, United States (Project 2020-8223).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (or Ethics Committee) of Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore of Rome, Italy (Study ID: 3226). Additionally, if applicable, the respective local ethics committee of each participating institution might have approved the EPICOVIDEHA.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study when required by the respective Ethics Committee.

Data Availability Statement

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

Conflicts of Interest

The funder of the study had no role in study design, data analysis, interpretation, or writing of the report. All authors had full access to the data and had final responsibility for the decision to submit for publication.

References

  1. Pagano, L.; Salmanton-García, J.; Marchesi, F.; Busca, A.; Corradini, P.; Hoenigl, M.; Klimko, N.; Koehler, P.; Pagliuca, A.; Passamonti, F.; et al. COVID-19 infection in adult patients with hematological malignancies: A European Hematology Association Survey (EPICOVIDEHA). J. Hematol. Oncol. 2021, 14, 168. [Google Scholar] [CrossRef] [PubMed]
  2. García-Suárez, J.; de la Cruz, J.; Cedillo, Á.; Llamas, P.; Duarte, R.; Jiménez-Yuste, V.; Hernández-Rivas, J.Á.; Gil-Manso, R.; Kwon, M.; Sánchez-Godoy, P.; et al. Impact of hematologic malignancy and type of cancer therapy on COVID-19 severity and mortality: Lessons from a large population-based registry study. J. Hematol. Oncol. 2020, 13, 133. [Google Scholar] [CrossRef] [PubMed]
  3. Cattaneo, C.; Daffini, R.; Pagani, C.; Salvetti, M.; Mancini, V.; Borlenghi, E.; D’Adda, M.; Oberti, M.; Paini, A.; De Ciuceis, C.; et al. Clinical characteristics and risk factors for mortality in hematologic patients affected by COVID-19. Cancer 2020, 126, 5069–5076. [Google Scholar] [CrossRef] [PubMed]
  4. Zeidan, A.M.; Boddu, P.C.; Patnaik, M.M.; Bewersdorf, J.P.; Stahl, M.; Rampal, R.K.; Shallis, R.; Steensma, D.P.; Savona, M.R.; Sekeres, M.A.; et al. Special considerations in the management of adult patients with acute leukemias and myeloid neoplasms in the COVID-19 era: Recommendations from a panel of international experts. Lancet Haematol. 2020, 7, e601–e612. [Google Scholar] [CrossRef]
  5. Marchesi, F.; Salmanton-García, J.; Emarah, Z.; Piukovics, K.; Nucci, M.; López-García, A.; Ráčil, Z.; Farina, F.; Popova, M.; Zompi, S.; et al. COVID-19 in adult acute myeloid leukemia patients: A long-term followup study from the European Hematology Association survey (EPICOVIDEHA). Haematologica, 2022; online ahead of print. [Google Scholar] [CrossRef]
  6. Hungria, V.; Garnica, M.; Crusoé, E.D.Q.; Filho, R.J.P.D.M.; Martinez, G.; Bittencourt, R.; de Farias, D.L.C.; Braga, W.M.; Neto, J.V.P.; Ribeiro, G.N.; et al. Managing patients with multiple myeloma during the COVID-19 pandemic: Recommendations from an expert panel–ABHH monoclonal gammopathies committe. Hematol. Transfus. Cell Ther. 2020, 42, 200–205. [Google Scholar] [CrossRef] [PubMed]
  7. Grever, M.; Andritsos, L.; Banerji, V.; Barrientos, J.C.; Bhat, S.; Blachly, J.S.; Call, T.; Cross, M.; Dearden, C.; Demeter, J.; et al. Hairy cell leukemia and COVID-19 adaptation of treatment guidelines. Leukemia 2021, 35, 1864–1872. [Google Scholar] [CrossRef]
  8. Lamure, S.; Salmanton-García, J.; Marieton, E.R.; Jaksic, O.; Kohn, M.; Marchesi, F.; Marchetti, M.; El-Ashwah, S.; Demirkan, F.; Valković, T.; et al. COVID-19 and Hairy-Cell Leukemia: An EPICOVIDEHA Survey. Blood Adv. 2022, 6, 3870–3874. [Google Scholar] [CrossRef]
  9. Taurino, D.; Frigeni, M.; Grassi, A.; Cavallaro, G.; Salmoiraghi, S.; Spinelli, O.; Rambaldi, A.; Lussana, F. Concurrent diagnosis of acute myeloid leukemia and symptomatic COVID-19 infection: A case report successfully treated with Azacitidine-Venetoclax combination. Mediterr. J. Hematol. Infect. Dis. 2021, 13, e2021057. [Google Scholar] [CrossRef]
  10. Orf, K.; Rogosic, S.; Dexter, D.; Ancliff, P.; Badle, S.; Brierley, J.; Cheng, D.; Dalton, C.; Dixon, G.; Du Pré, P.; et al. Remdesivir during induction chemotherapy for newly diagnosed paediatric acute lymphoblastic leukaemia with concomitant SARS-CoV-2 infection. Br. J. Haematol. 2020, 190, e274–e276. [Google Scholar] [CrossRef]
  11. Ghandili, S.; Pfefferle, S.; Roedl, K.; Sonnemann, P.; Karagiannis, P.; Boenisch, O.; Kluge, S.; Schmiedel, S.; Ittrich, H.; Rohde, H.; et al. Challenges in treatment of patients with acute leukemia and COVID-19: A series of 12 patients. Blood Adv. 2020, 4, 5936–5941. [Google Scholar] [CrossRef]
  12. Salmanton-García, J.; Busca, A.; Cornely, O.A.; Corradini, P.; Hoenigl, M.; Klimko, N.; Marchesi, F.; Pagliuca, A.; Passamonti, F.; Koehler, P.; et al. EPICOVIDEHA: A Ready to Use Platform for Epidemiological Studies in Hematological Patients With COVID-19. HemaSphere 2021, 5, e612. [Google Scholar] [CrossRef] [PubMed]
  13. National Cancer Institute Dictionary of Cancer Terms. Available online: https://www.cancer.gov/publications/dictionaries/cancer-terms (accessed on 1 May 2022).
  14. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Available online: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm (accessed on 1 May 2022).
  15. Palanques-Pastor, T.; Megías-Vericat, J.E.; Martínez, P.; López Lorenzo, J.L.; Cornago Navascués, J.; Rodriguez Macias, G.; Cano, I.; Arnan Sangerman, M.; Vidriales Vicente, M.B.; Algarra Algarra, J.L.; et al. Characteristics, clinical outcomes, and risk factors of SARS-COV-2 infection in adult acute myeloid leukemia patients: Experience of the PETHEMA group. Leuk. Lymphoma 2021, 62, 2928–2938. [Google Scholar] [CrossRef] [PubMed]
  16. Ribera, J.-M.; Morgades, M.; Coll, R.; Barba, P.; López-Lorenzo, J.-L.; Montesinos, P.; Foncillas, M.-A.; Cabrero, M.; Gómez-Centurión, I.; Morales, M.-D.; et al. Frequency, Clinical Characteristics and Outcome of Adults With Acute Lymphoblastic Leukemia and COVID 19 Infection in the First vs. Second Pandemic Wave in Spain. Clin. Lymphoma Myeloma Leuk. 2021, 21, e801–e809. [Google Scholar] [CrossRef] [PubMed]
  17. Chiaretti, S.; Bonifacio, M.; Agrippino, R.; Giglio, F.; Annunziata, M.; Curti, A.; Del Principe, M.I.; Salutari, P.; Sciumè, M.; Delia, M.; et al. COVID-19 infection in acute lymphoblastic leukemia over 15 months of the pandemic. A Campus ALL report. Haematologica 2022, 107, 1955–1959. [Google Scholar] [CrossRef]
  18. Yarza, R.; Bover, M.; Paredes, D.; López-López, F.; Jara-Casas, D.; Castelo-Loureiro, A.; Baena, J.; Mazarico, J.M.; Folgueira, M.D.; Meléndez-Carmona, M.Á.; 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, 242–250. [Google Scholar] [CrossRef]
  19. Ljungman, P.; de la Camara, R.; Mikulska, M.; Tridello, G.; Aguado, B.; Zahrani, M.A.; Apperley, J.; Berceanu, A.; Bofarull, R.M.; Calbacho, M.; et al. COVID-19 and stem cell transplantation; results from an EBMT and GETH multicentre prospective survey. Leukemia 2021, 35, 2885–2894. [Google Scholar] [CrossRef]
  20. Piñana, J.L.; Martino, R.; García-García, I.; Parody, R.; Morales, M.D.; Benzo, G.; Gómez-Catalan, I.; Coll, R.; De La Fuente, I.; Luna, A.; et al. Risk factors and outcome of COVID-19 in patients with haematological malignancies. Exp. Hematol. Oncol. 2020, 9, 21. [Google Scholar] [CrossRef]
  21. Stahl, M.; Narendra, V.; Jee, J.; Derkach, A.; Maloy, M.; Geyer, M.B.; Mato, A.R.; Roeker, L.E.; Tallman, M.S.; Shah, G.L.; et al. Neutropenia in adult acute myeloid leukemia patients represents a powerful risk factor for COVID-19 related mortality. Leuk. Lymphoma 2021, 62, 1940–1948. [Google Scholar] [CrossRef]
  22. Cesaro, S.; Ljungman, P.; Mikulska, M.; Hirsch, H.H.; von Lilienfeld-Toal, M.; Cordonnier, C.; Meylan, S.; Mehra, V.; Styczynski, J.; Marchesi, F.; et al. Recommendations for the management of COVID-19 in patients with haematological malignancies or haematopoietic cell transplantation, from the 2021 European Conference on Infections in Leukaemia (ECIL 9). Leukemia 2022, 36, 1467–1480. [Google Scholar] [CrossRef]
  23. Salmanton-García, J.; Sprute, R.; Stemler, J.; Bartoletti, M.; Dupont, D.; Valerio, M.; García-Vidal, C.; Falces-Romero, I.; Machado, M.; de la Villa, S.; et al. COVID-19–Associated Pulmonary Aspergillosis, March–August 2020. Emerg. Infect. Dis. 2021, 27, 1077–1086. [Google Scholar] [CrossRef]
  24. Prattes, J.; Wauters, J.; Giacobbe, D.R.; Salmanton-García, J.; Maertens, J.; Bourgeois, M.; Reynders, M.; Rutsaert, L.; Van Regenmortel, N.; Lormans, P.; et al. Risk factors and outcome of pulmonary aspergillosis in critically ill coronavirus disease 2019 patients—A multinational observational study by the European Confederation of Medical Mycology. Clin. Microbiol. Infect. 2022, 28, 580–587. [Google Scholar] [CrossRef] [PubMed]
  25. Zappasodi, P.; Cattaneo, C.; Ferretti, V.; Mina, R.; Ferreri, A.J.M.; Merli, F.; Oberti, M.; Krampera, M.; Romano, A.; Zerbi, C.; et al. Secondary infections worsen the outcome of COVID-19 in patients with hematological malignancies a report from the ITA-HEMA-COV. Hematol. Oncol. 2022; online ahead of print. [Google Scholar] [CrossRef]
  26. Cattaneo, C.; Pagani, C.; Cancelli, V.; Imberti, L.; Roccaro, A.M.; Notarangelo, L.D.; Rossi, G. Reduction in the rate and improvement in the prognosis of COVID-19 in haematological patients over time. Leukemia 2021, 35, 632–634. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overall survival probability. (a) Cohort. (b) Patients by malignancy.
Figure 1. Overall survival probability. (a) Cohort. (b) Patients by malignancy.
Cancers 14 05530 g001aCancers 14 05530 g001b
Figure 2. 30-day survival probability of treated vs. untreated patients.
Figure 2. 30-day survival probability of treated vs. untreated patients.
Cancers 14 05530 g002
Table 1. Characteristics of patients with simultaneous onset of haematological malignancy and COVID-19.
Table 1. Characteristics of patients with simultaneous onset of haematological malignancy and COVID-19.
n%
Sex
Female18641.3%
Male26458.7%
Age, median (IQR) [absolute range]65 (53 to 75) [18 to 95]
18–25 years old153.3%
26–50 years old8418.7%
51–69 years old17438.7%
≥70 years old17739.3%
Comorbidities before COVID-19
No comorbidities16837.3%
1 comorbidity14031.1%
2 comorbidities8118%
3 or more comorbidities6113.6%
  Chronic cardiopathy15634.7%
  Chronic pulmonary disease6113.6%
  Diabetes mellitus7717.1%
  Liver disease184%
  Obesity439.6%
  Renal impairment439.6%
  Smoking history5211.6%
  No risk factor identified16837.3%
Baseline malignancy
Days between malignancy diagnosis and COVID-19 diagnosis, median (IQR) [absolute range] −11 (−21 to −2) [−33 to 3]
Acute lymphoid leukaemia327.1%
Chronic lymphoid leukaemia276%
Acute myeloid leukaemia12928.7%
Chronic myeloid leukaemia92%
Hodgkin lymphoma163.6%
Non-Hodgkin lymphoma14231.6%
  Indolent429.3%
  Aggressive9120.2%
  Unknown71.6%
Essential thrombocythemia20.4%
Multiple myeloma5512.2%
Myelodysplastic syndrome255.6%
Myelofibrosis30.7%
Hairy cell leukaemia102.2%
Neutrophils at COVID-19 diagnosis
≤5006915.3%
501–999286.2%
≥100030768.2%
Lymphocytes at COVID-19 diagnosis
≤2004710.4%
201–4995412%
≥50031570%
COVID-19 infection
Asymptomatic10623.6%
Mild infection5812.9%
Severe infection17037.8%
Critical infection11625.8%
COVID-19 symptoms at infection onset
Pulmonary15133.6%
Pulmonary + extrapulmonary10623.6%
Extrapulmonary6614.7%
Screening12728.2%
COVID-19 vaccination
Not vaccinated36080%
One dose153.3%
Two doses4610.2%
Three doses296.4%
COVID-19 treatment
No specific treatment reported27761.6%
Antivirals + monoclonal antibodies ± corticosteroids ± plasma81.8%
Antivirals ± corticosteroids ± plasma5211.6%
Corticosteroids7216%
Monoclonal antibodies ± plasma ± corticosteroids316.9%
Plasma ± corticosteroids102.2%
Stay during COVID-19 episode
Hospital38084.4%
  Days of the stay in ICU, median (IQR) [absolute range]15 (7 to 28) [1 to 150]
  ICU11625.8%
    Days of the stay in ICU, median (IQR) [absolute range]8 (4 to 20) [1 to 56]
    Invasive mechanical ventilation7416.4%
    Non-invasive mechanical ventilation429.3%
Home9320.7%
Negative days indicate days before COVID-19 diagnosis. Positive days indicate days after COVID-19 diagnosis. Day of COVID-19 diagnosis is set as day 0. Asymptomatic: no clinical signs or symptoms; mild: non-pneumonia and mild pneumonia; severe: dyspnea, respiratory frequency ≥ 30 breaths per min, SpO2 ≤ 93%, PaO2/FiO2 < 300, or lung infiltrates > 50%); critical: patients admitted in intensive care for respiratory failure, septic shock, or multiple organ dysfunction or failure. COVID-19, coronavirus disease 2019; ICU, intensive care unit; IQR, interquartile range.
Table 2. Clinical characteristics of treated and not treated patients.
Table 2. Clinical characteristics of treated and not treated patients.
No HM Tx at AllHM Tx at Least after COVID-19 dxp ValueNo HM Tx at AllHM Txp Value
n%n%n%n%
Sex 0.031 0.021
Female3431.8%9644.2%3431.8%15244.3%
Male7368.2%12155.8%7368.2%19155.7%
Age, median (IQR) [absolute range]68 (56–79)
[24–92]
61 (47–72)
[18–95]
<0.00168 (56–79)
[24–92]
64 (50–74)
[18–95]
0.003
Comorbidities Before COVID-19 0.031 0.093
No comorbidities3734.6%8639.6%3734.6%13138.2%
1 comorbidity2927.1%7132.7%2927.1%11132.4%
2 comorbidities2826.2%2812.9%2826.2%5315.5%
3 or more comorbidities1312.1%3214.7%1312.1%4814.0%
Baseline malignancy <0.001 <0.001
ALL43.7%198.8%43.7%288.2%
AML1715.9%6228.6%1715.9%11232.7%
Lymphoma3229.9%8237.8%3229.9%12436.2%
CLD2624.3%94.1%2624.3%133.8%
MM1211.2%2913.4%1211.2%4312.5%
MDS/MPS1615.0%167.4%1615.0%236.7%
Anti-SARS-CoV-2 vaccination2624.3%3817.5%0.1492624.3%6418.7%0.203
COVID-19 severity 0.019 0.092
Asymptomatic2018.7%6429.5%2018.7%8625.1%
Mild infection1110.3%2712.4%1110.3%4713.7%
Severe infection3936.4%8338.2%3936.4%13138.2%
Critical infection3734.6%4319.8%3734.6%7923.0%
COVID-19 diagnostic period <0.001 0.001
March 2020–September 20202725.2%5625.8%2725.2%9126.5%
October 2020–February 20212523.4%9945.6%2523.4%14241.4%
March 2021–November 20212422.4%2913.4%2422.4%4713.7%
December 2021–February 20223129.0%3315.2%3129.0%6318.4%
Any COVID-19 treatment4643.0%6630.4%0.0254643.0%12737.0%0.268
Day-30 mortality4542.1%3315.2%<0.0014542.1%9427.4%0.004
ALL, acute lymphoid leukaemia; AML, acute myeloid leukaemia; CLD, chronic lymphoproliferative disorder; COVID-19, coronavirus disease 2019; Dx, diagnosis; HM, haematological malignancy; IQR, interquartile range; MDS, myelodysplastic syndrome; MM, multiple myeloma; MPS, myeloproliferative syndrome; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; Tx, treatment.
Table 3. Univariable and multivariable analysis for risk factors for mortality (whole cohort).
Table 3. Univariable and multivariable analysis for risk factors for mortality (whole cohort).
UnivariableMultivariable
p ValueHR95% CIp ValueHR95% CI
LowerUpperLowerUpper
Sex
Female----
Male0.9650.9930.7351.342
Age<0.0011.0351.0241.046<0.0011.0331.0191.047
Baseline malignancy
ALL--------
AML0.0152.6321.2105.7260.0802.3070.9065.873
Lymphoma0.6561.1980.5412.6500.9570.9740.3692.567
CLD0.8321.1080.4302.8600.0820.3650.1171.137
MM0.0852.0900.9044.8330.9411.0400.3742.889
MDS/MPS0.2451.7040.6944.1800.7520.8380.2822.495
COVID-19 infection severity
Asymptomatic--------
Mild infection0.9191.0360.5252.0450.8621.0680.5092.239
Severe infection0.0801.5450.9492.5150.0261.8041.0743.032
Critical infection<0.0014.5982.8967.299<0.0015.5233.3289.166
Comorbidities at COVID-19 onset
No comorbidities--------
1 comorbidity0.0331.5271.0342.2560.6181.1130.7321.692
2 comorbidities<0.0012.6301.7463.9600.0151.7671.1192.793
3 or more comorbidities0.0141.8171.1292.9240.0661.6800.9672.918
Season COVID-19 diagnosis
March 2020–September 2020--------
October 2020–February 20210.0030.6030.4300.8450.6991.0840.7201.634
March 2021–November 20210.0600.6390.4001.0190.4021.2800.7192.279
December 2021–February 2022<0.0010.3840.2220.6660.7280.8800.4281.810
Malignancy treatment?
No----
Before COVID-19 diagnosis0.0530.7170.5121.004
After COVID-19 diagnosis<0.0010.2510.1540.408
Neutrophils at COVID-19 onset
≤500----
501–9990.6500.8580.4421.665
≥10000.0820.7060.4751.045
Lymphocytes at COVID-19 onset
≤200--------
201–4990.0580.5820.3331.0180.2100.6830.3771.239
≥5000.0050.5530.3660.8360.0240.5730.3520.931
Chemotherapy induced neutropenia
No--------
Yes0.0710.7220.5061.0290.2391.2850.8461.950
No treatment administered0.0111.6021.1122.308<0.0013.4492.2245.349
Secondary infection after COVID-190.1250.7400.5031.087
COVID-19 treatment
No specific treatment reported--------
Antivirals + monoclonal antibodies ± corticosteroids ± plasma0.2130.2860.0402.0490.1050.1900.0251.414
Antivirals ± corticosteroids ± plasma0.7931.0640.6681.6950.3950.7980.4761.341
Corticosteroids0.7910.9440.6171.4440.0330.6050.3810.961
Monoclonal antibodies ± plasma ± corticosteroids0.0260.3600.1470.8820.0810.4420.1771.107
Plasma ± corticosteroids0.9051.0630.3922.8800.2690.5570.1981.571
ALL, acute lymphoid leukaemia; AML, acute myeloid leukaemia; CI, confidence interval; CLD, chronic lymphoproliferative disorder; COVID-19, coronavirus disease 2019; HR, hazard ratio; MDS, myelodysplastic syndrome; MM, multiple myeloma; MPS, myeloproliferative syndrome asymptomatic: no clinical signs or symptoms; mild: non-pneumonia and mild pneumonia; severe: dyspnoea, respiratory frequency ≥ 30 breaths per min, SpO2 ≤ 93%, PaO2/FiO2 < 300, or lung infiltrates > 50%); critical: patients admitted in intensive care for respiratory failure, septic shock, or multiple organ dysfunction or failure.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cattaneo, C.; Salmanton-García, J.; Marchesi, F.; El-Ashwah, S.; Itri, F.; Weinbergerová, B.; Gomes Da Silva, M.; Dargenio, M.; Dávila-Valls, J.; Martín-Pérez, S.; et al. Simultaneous Onset of Haematological Malignancy and COVID: An Epicovideha Survey. Cancers 2022, 14, 5530. https://doi.org/10.3390/cancers14225530

AMA Style

Cattaneo C, Salmanton-García J, Marchesi F, El-Ashwah S, Itri F, Weinbergerová B, Gomes Da Silva M, Dargenio M, Dávila-Valls J, Martín-Pérez S, et al. Simultaneous Onset of Haematological Malignancy and COVID: An Epicovideha Survey. Cancers. 2022; 14(22):5530. https://doi.org/10.3390/cancers14225530

Chicago/Turabian Style

Cattaneo, Chiara, Jon Salmanton-García, Francesco Marchesi, Shaimaa El-Ashwah, Federico Itri, Barbora Weinbergerová, Maria Gomes Da Silva, Michelina Dargenio, Julio Dávila-Valls, Sonia Martín-Pérez, and et al. 2022. "Simultaneous Onset of Haematological Malignancy and COVID: An Epicovideha Survey" Cancers 14, no. 22: 5530. https://doi.org/10.3390/cancers14225530

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