Treatment Approach, Pharmacological Agents and Vaccines


Acknowledgment

We acknowledge the contribution by Dr. Mayur Ramesh of the Infection Diseases Service at Henry Ford Hospital, Detroit, Michigan, USA.

Treatment Approach for the Hospitalized Patient With COVID-19

Introduction

The goal of this chapter is to inform the neurologist of the disease-specific treatments that exist and are recommended for the patient with COVID-19. The proposed approach to management is tiered according to the severity of illness and encompasses medications that are recommended by several notable organizations, such as the National Institute of Health (NIH) and the Society of Critical Care Medicine (SCCM), according to their most recent guidelines. Treatment of severe COVID-19 is complex and requires expertise in critical care medicine including hemodynamic and ventilatory support, topics that are beyond the scope of this book. An appendix grouping several medications related to the care of patients with COVID-19 is provided at the end of this book (Appendix: Medication). We also present a summary table of the various treatments with a particular mention of the neurological complications and considerations for each one of them ( Table 12.1 ).

Table 12.1
Selected COVID-19-Specific Treatments in Use in the United States and Other Countries.
Name Mechanism of Action Efficacy Safety Neurologic Considerations
Bamlanivimab + etesevimab Bamlanivimab (also known as LY-CoV555 and LY3819253) is a neutralizing monoclonal antibody that targets the RBD of the S protein of SARS-CoV-2.
Etesevimab (also known as LY-CoV016 and LY3832479) is another neutralizing monoclonal antibody that binds to a different but overlapping epitope in the RBD of the SARS-CoV-2 S protein.
Authorized for use in outpatient cases at high risk of progression to severe COVID-19. 70% relative risk reduction of hospitalization or death compared to placebo.
Not authorized for hospitalized patients or those requiring oxygen supplementation except in special circumstances.
Associated with few and mainly low-grade toxic effects.
BLAZE-1 trial data demonstrated nausea, diarrhea, dizziness, and headaches as most common side effects. 1% of patients experienced hypersensitivity reactions.
No significant reported neurologic complications or considerations to date.
Baricitinib Selective inhibitor of JAK-1 and − 2 and causes inhibition of the intracellular signaling pathway of cytokines known to be elevated in severe Covid-19 including IL-2 and IL-6. ACTT-2 Trial compared Baricitinib + remdesivir to remdesivir alone with 30% improvement in time to recovery and clinical status in the combination group. Results were more positive in patients receiving high-flow oxygen and noninvasive BIPAP. Adverse events were less common in combination therapy arm than in the remdesivir alone arm.
Most common side effects include upper respiratory infections, headaches, and nasopharyngitis. Dose-related neutropenia and lymphopenia have been observed.
No major neurologic complications reported.
20% CNS penetration (higher than most other reported COVID-19 treatments)
Casirivimab + imdevimab Casirivimab (previously REGN10933) and imdevimab (previously REGN10987) are recombinant human monoclonal antibodies that bind to nonoverlapping epitopes of the S protein RBD of SARS-CoV-2. Available through FDA emergency use authorization for mild-moderate COVID-19 cases at high risk of progression to severe disease. 71% relative risk reduction of hospitalization or death compared to placebo.
Not authorized for hospital patients or those requiring oxygen supplementation except in special circumstances.
Associated with few and mainly low-grade toxic effects.
Percentages of patients with hypersensitivity reactions, infusion-related reactions, and other adverse events were similar in the combined REGN-COV2 dose groups and the placebo group.
No significant reported neurologic complications or considerations to date.
Convalescent Plasma Pooled human IgG antibodies from COVID-19 survivors contain IgG with a high titer (1:1000) of anti-SARS-CoV-2 S protein 15% reduction in progression to severe COVID-19 when given in the first 72-h of symptom onset. No major adverse events in largest COVID-19 trial, although transfusions may be associated with fluid overload, transfusion associated lung injury (TRALI), and allergic reaction No significant reported neurologic complications or considerations to date.
Corticosteroids Physiologic activity to reduce systemic inflammatory responses via effects on cytokine production, potentially reducing inflammation-related lung damage Reduction in death rates when administered to hospital patients receiving supplemental O 2 or mechanical ventilation (RECOVERY trial). High doses associated with metabolic derangement, increased infection risk, increased viral replication, delayed viral clearance. Protracted use can contribute to myopathy, neuromuscular weakness, delirium, and psychiatric symptoms.
Hydroxychloroquine/ chloroquine Impacts viral entry into cells and effects viral protein synthesis through multiple pathways. Conflicting results; most randomized studies conclude no clinical benefit when HCQ is added to standard of care. Not recommended for treatment of inpatient or outpatient COVID-19. Narrow therapeutic index; can cause QT prolongation, torsade de pointes, bone marrow suppression, seizure, retinopathy, and myopathy. Mood disorders, psychosis.
At high doses can cause ototoxicity leading to hearing loss, tinnitus, and imbalance.
21% CNS penetration.
Immunoglobulin (IVIG) Pooled human IgG antibodies consisting of Fab fragment and Fc fragment which aid in antigen recognition and immune system response.
Used in a variety of autoimmune and inflammatory disorders as well as tested in bacterial, viral and fungal infections.
Significant reduction of in-hospital mortality in small randomized, controlled trials of IVIG versus placebo.
Has been studied specifically in lymphopenic patients
Risks similar to the expected risks of plasma transfusion and transfusion reactions. Infusions must be done slowly over several days. Headache and dizziness.
Thromboembolic events (TIA, stroke, CVST).
Rare reports of aseptic meningitis and acute encephalopathy following IVIG infusion have been suggested from immunoallergic reaction from entry of IgG into CSF spaces
Ivermectin Inhibits viral replication; also used in strongyloides and onchocerca infections Decreases SARS-CoV-2 replication in vitro, but failed to improve time to resolution of symptoms in humans Common side effects include rash, muscle aches, headache, fever Dizziness, vertigo, tremor, lethargy.
Encephalopathy, stupor or coma in individuals with mdr-1 gene mutations or when combined with CYP-3A4 inhibitors.
Lopinavir +/− ritonavir HIV-1 protease inhibitor + ritonavir booster (inhibits lopinavir metabolism). Also effective in treating SARS-CoV. Some reports of efficacy in individual cases, but more studies are needed. Most common side effects include renal tubular acidosis, AKI, dermatitis. Used in a case of COVID-19-associated cerebritis with significant improvement.
0.02% CNS penetration.
Remdesivir Adenosine analog, incorporates into viral RNA, disrupts transcription Shortens time to recovery when administered within 1 day of symptom onset Associated with AKI or transaminitis No neurologic adverse events reported.
<5% CNS penetration.
Tocilizumab Recombinant anti-IL-6 receptor monoclonal antibody which can limit cytokine-related pulmonary injury from cytokine storm.
Used in treatment of rheumatoid arthritis.
Has demonstrated efficacy in improving fevers, oxygen requirements, and CT findings in patients with severe COVID-19 pneumonia. 12% reduction in mortality in meta-analysis of use in COVID-19 patients. Associated with infections (skin, cellulitis), gastrointestinal disorders, and transfusion reactions. Few case reports of cerebral microangiopathy and leukoencephalopathy when used in patients with pre-existing rheumatologic conditions.
0.1% CNS penetration

Treatment Approach for Patients With COVID-19

The approach more commonly adopted to caring for the hospitalized patient with COVID-19 depends on the degree of severity of the illness. Therefore defining severity in COVID-19 is important and one of the first steps in managing such patients. Dyspnea is the key in categorizing patients with suspected or confirmed COVID-19. Patients with no dyspnea but with other symptoms such as fever, chills, cough, malaise, anosmia, headaches, and upper respiratory symptoms are considered nonsevere and rarely require hospitalization. Once dyspnea is observed and the patient requires supplemental oxygen, the patient’s condition is deemed severe. The Surviving Sepsis Campaign Guidelines on the Management of Adults with Coronavirus Disease 2019 (COVID-19) in the ICU , for instance, further differentiates severe from critical disease. Severe disease concerns patients with clinical signs of pneumonia (e.g., tachypnea, dyspnea, fever, and cough) and one of the following: respiratory rate greater than 30 breaths per minute, severe respiratory distress, or oxygen saturation of less than 90% on room air. Critical disease identifies patients with acute respiratory distress syndrome (ARDS) or respiratory distress requiring mechanical ventilation, sepsis, or septic shock. The NIH Panel on COVID-19 Treatment Guidelines published a pharmacological management based on disease severity, which can be found in Fig. 12.1 .

Fig. 12.1, Pharmacological management guide of patients with COVID-19 based on disease severity.

In general terms:

  • 1.

    Only supportive treatment and monitoring is recommended for patients with nonsevere disease.

  • 2.

    Anti-SARS-CoV-2 monoclonal antibody combinations, such as bamlanivimab plus etesevimab, or casirivimab plus imdevimab, should be considered for nonhospitalized patients or patients with mild COVID-19 hospitalized for reasons other than COVID-19 who are at high risk of disease progression.

  • 3.

    Dexamethasone is strongly recommended in patients who require any amount of supplemental oxygen and/or mechanical ventilation or extracorporeal membrane oxygenation (ECMO).

  • 4.

    Remdesivir is the only FDA-approved medication for the treatment of COVID-19. It is recommended in patients on supplemental oxygen but not in patients who present to the hospital needing mechanical ventilation or ECMO, since their condition is considered too advanced to derive any significant benefit from the antiviral. The combination dexamethasone/remdesivir has not been studied in patients requiring mechanical ventilation.

  • 5.

    Interleukin-6 antagonists, such as tocilizumab, can be added to dexamethasone in patients who display evidence of systemic inflammation or whose respiratory condition rapidly decompensates, requiring escalating levels of oxygen, increasing settings on noninvasive ventilation, or mechanical ventilation.

Importantly, several societies and organizations now recommend against the use of hydroxychloroquine or chloroquine (with or without azithromycin), for either hospitalized or nonhospitalized patients with COVID-19. Also, due to poor-quality data, insufficient data, or lack of benefit, the use of convalescent plasma or intravenous immunoglobulin (IVIG) is not authorized for use in nonhospitalized patients. The NIH Panel on COVID-19 Treatment Guidelines recommends against its use in patients with severe disease who are mechanically ventilated, or those who are hospitalized but not on mechanical ventilation, except in the context of a clinical trial. It is however authorized under the EAU for use in hospitalized patients with immune deficiencies. Additionally, due to lack of evidence, the guidelines recommend neither for nor against the routine use of therapeutic anticoagulation for hospitalized patients with COVID-19 who do not have confirmed venous thromboembolic disease.

Special Considerations for COVID-19-Specific Treatments in Patients With Neurological Conditions

In this section, we provide information about the neurological considerations of drugs used for the specific treatment of COVID-19. These drugs and a summary of their neurotoxicities can be found in Table 12.1 . We hope that neurologists consulting on hospitalized patients with COVID-19 will find this data relevant and useful.

Corticosteroids . Corticosteroids can have a plethora of complications depending on their route of administration, dose, and duration of use. In addition to hyperglycemia which can worsen encephalopathy, delay wound healing and independently worsen outcomes in patients with stroke or cerebral edema, these drugs have other more direct effects on central and peripheral nervous systems. Mood disturbances are the most pervasive and can occur with a few days of treatment, usually causing early euphoria and anxiety. Depression can set in with more prolonged therapy. Memory impairment also occurs with more protracted treatment and usually happens with increased frequency and more rapidly in elderly patients (3 months as opposed to 1 year). Psychosis also tends to happen with prolonged therapy and is therefore not expected in steroid therapy used for COVID-19 infection. Delirium, sleep disturbances, and akathisia (psychomotor restlessness) are typical central nervous side effects of corticosteroids. In addition to their central complications, steroids can lead to ICU-acquired weakness, ICUAW (also known as critical care myopathy and neuropathy), especially when coadministered with neuromuscular blocking agents, a combination that is commonly used for the treatment of severe ARDS in patients with COVID-19. The association between steroids and ICUAW is almost twofold. Other important risk factors for ICUAW are mechanical ventilation, prolonged ICU stay, and sepsis and septic shock. Finally, although a ubiquitous treatment for it, steroids can initially worsen a myasthenic crisis and need to be started or increased with caution in patients with a confirmed or suspected diagnosis of MG. IVIG or plasmapheresis may be needed as a bridge to steroids when the symptoms get worse with treatment.

Hydroxychloroquine/chloroquine . The antimalarial drugs are associated with a high risk of retinopathy which is dose and duration dependent (7.5%). Ototoxicity in the form of sensorineural hearing loss, tinnitus, and imbalance can happen with both chloroquine and hydroxychloroquine and tends to happen more abruptly with the former and more insidiously with the latter. The ototoxicity risk at the doses used in COVID-19 infection is however not known. Furthermore, these drugs can also prolong the QT interval, making the choice of antipsychotics in delirious patients with COVID-19 more challenging. They have been linked to several psychiatric manifestations such as mania, depression, and psychosis as well as central nervous side effects like insomnia, dizziness, and headache in 4%–6% of patients. Although seizure is listed in the package insert for both drugs, it is a very rare occurrence and data is limited to case reports and case series, without any causal relationship firmly established.

Immunoglobulins . Thromboembolic events after IVIG can occur within hours to days of treatment for arterial clots and days to weeks for venous ones. They can lead to acute coronary syndromes, transient ischemic attacks (TIA) and stroke, cerebral venous sinus thrombosis, pulmonary embolism, and deep venous thrombosis (DVT). Adequate hydration, slowing down the rate or breaking down the infusion into several smaller doses can help prevent this complication. Discontinuing the treatment is likely the safest approach in cases of thromboembolic events. Aseptic meningitis is another neurological complication of IVIG. It occurs in about 1% of patients and its pathophysiology is poorly understood. It is a self-limiting condition and resolves by slowing down the rate of infusion along with symptomatic care including use of proper hydration, analgesics, antipyretics, and antihistamines.

Ivermectin . The anthelmintic is not used or recommended for use in COVID-19 outside of clinical trials in the United States. It is used in countries like India, South Africa, Zimbabwe, or Mexico. This drug is known to cause dizziness (2.8%), vertigo, tremor, and lethargy (< 1%). Although coma and encephalopathy were reported in patients treated with ivermectin for Onchocerciasis volvulus in Africa, many of these cases were thought to be due to concomitant infection with Loa loa (loiasis). Cotreatment with drugs that inhibit the CYP3A4 enzyme can also predispose to higher neurotoxicity from ivermectin.

Tocilizumab. Headaches (7%) and dizziness (3%) are the most commonly reported nervous side effects of tocilizumab. A case of multifocal cerebral microangiopathy arising shortly after a single infusion of the medication in a patient with rheumatoid arthritis was reported and was believed to be an immune-mediated vasculitic process. Additionally, two cases of leukoencephalopathy in patients with rheumatoid arthritis treated with tocilizumab were reported. One of them was associated with limbic encephalitis and both cases showed symptoms several months following treatment with the IL-6 antagonist. These are believed to be exceedingly rare occurrences.

COVID-19 Vaccines

Introduction

Due to the rapid spread and devastating effects caused by SARS-CoV-2 on the human loss of life and livelihood, the need for vaccine development has been an unrivaled priority. Publication of the viral genome in January 2020 enabled many pharmaceutical companies around the world to start developing a variety of vaccines against the pathogen.

Vaccines have to go through a rigorous series of steps before being approved for marketing. After research, discovery, and development, the vaccine is tested in animals (preclinical stage). In all, 77 COVID-19 vaccines are currently undergoing animal trials. If the vaccine passes this first checkpoint, it proceeds to phase 1 clinical trials. In this phase, the safety of the vaccine using various doses is determined by testing it in a small group of healthy volunteers (20–100 subjects). There are currently 49 COVID-19 vaccines in phase 1 trials. Phase 2 trials are randomized, controlled, and recruit hundreds of patients at risk for developing the condition. The goal is to measure the vaccine’s immunogenicity (i.e., the ability of the vaccine to elicit an immune response) and determine its tolerability and safety. A total of 37 COVID-19 vaccines are currently in phase 2 trials. Phase 3 aims at gathering more information on the effectiveness of the vaccine and detecting less common side effects by recruiting a much larger cohort of subjects (usually, thousands) and randomizing them to vaccine or placebo arm. For vaccine approval, the United States FDA generally recommends that there be at least a 50% reduction in the primary endpoint of the phase 3 trial, which is usually effective in vaccine development. In all, 27 COVID-19 vaccines are presently in this third stage of the study. In the United States, three vaccines (BNT162b2, mRNA-1273, and Ad26.COV2·S) have received authorization for emergency use by the FDA but are not yet fully approved. Four vaccine trials were ultimately abandoned due to lack of effectiveness.

Further complicating the matter are the naturally occurring sporadic mutations of the virus leading to regional variants that are more transmissible, evade the traditional detection assays, and make vaccine development more challenging. Among the many SARS-CoV-2 virus mutants present across the world, five are of specific concern in the United States because they lead to more severe disease, greater transmissibility, and decreased neutralization by antibodies acquired via infection or vaccination: the United Kingdom, the South African, the Brazilian, and the two California variants.

Of note, none of these vaccines has been studied in human pregnancy or children, although some trials are currently underway in these populations. Long-term effects are still not known due to a lack of data but are presumed to be few. All currently used vaccines are in the form of intramuscular injection although there are several nasal mist vaccines in development.

At the time of writing, at the end of the first week of May 2021, approximately 257 million vaccines have been administered in the United States, with 34% of the population being fully vaccinated and 46% having received at least one dose. Pfizer-BioNTech’s vaccine leads in the number of doses administered followed closely by Moderna’s mRNA-1273. Johnson & Johnson/Janssen’s Ad26.COV2·S vaccine trails behind others due to the later issuance of emergency use authorization by the FDA.

Types of SARS-CoV-2 Vaccines

Most human antibodies produced in response to SARS-CoV-2 are in the form of neutralizing antibodies directed against the receptor-binding domain of the spike protein, which is located on the surface of the viral envelope and acts as the portal of viral entry into the host cells ( Chapter 2 ). Variants of SARS-CoV-2 generally carry a mutation in the receptor-binding domain of the spike protein. Although most mutations make the virus less infectious, some of them, like D614G present in all the variants of interest mentioned above, have been shown to increase the virus’ affinity to the ACE2 receptor, evade the host’s neutralizing antibodies, and jeopardize vaccine effectiveness since the vast majority of vaccines target the spike protein.

Vaccines currently developed for COVID-19 come in different flavors, such as mRNA, adenoviral vector, protein subunit, DNA, or inactivated viruses. Each vaccine type is associated with a different mechanism of action, delivery method, advantages, and disadvantages. Here, we give a brief overview of the various types along with a description of a few pharmaceutical products in use around the world for each category. Table 12.2 gives a summary of the efficacy, regimen, and neurological complications of some of the marketed vaccines in use around the world.

Table 12.2
Selected COVID-19 Vaccines With Emergency Use Authorization Around the World.
Name Company Vaccine Type Reported Efficacy (%) Regimen Notable Neurological Side Effects a
BNT162b2 Pfizer/BioNTech mRNA 95 2 doses; 3 weeks apart
mRNA-1273 Moderna mRNA 94.5 2 doses; 4 weeks apart
Ad26.COV2·S Johnson & Johnson/ Janssen Adenovirus vector 61–72 1 dose CVST (VITT/TTS)
Sputnik V Gamaleya Research Institute of Epidemiology and Microbiology Adenovirus vector 91.6 2 doses; 3 weeks apart
AZD1222 Oxford/ Astrazeneca Adenovirus vector 79 2 doses; 8–12 weeks apart
  • -

    CVST (VITT/TTS)

  • -

    Transverse myelitis (1 case)

ZF2001 Anhui Zhifei/ Longcom
Institute of Medical Biology at the Chinese Academy of Medical Sciences
Protein subunit Unknown 3 doses; 4 weeks apart
BBIBP-CorV Sinopharm Attenuated 79.34 2 doses; 3 weeks apart
CoronaVac Sinovac Attenuated 50.38–83.5 2 doses; 2 weeks apart
BBV152 Bharat Biotech Attenuated 80.6 2 doses; 4 weeks apart

a Only those neurological side effects that were probably related to vaccination are noted here. No causal relationship was found between the reported cases of Bell’s palsy in mRNA vaccine recipients, or GBS in J&J/Janssen vaccine recipients. Dizziness and headaches are mild neurological side effects reported with many of the vaccines included in this table.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here