Article Type
Changed
Mon, 09/30/2024 - 14:47

Unlike conventional immunosuppression, treatments approved since 2017 are giving patients with myasthenia gravis targeted options to better match their needs, desires, and tolerance. Used appropriately, newer treatments can provide dramatic results faster and more safely than broad immunosuppressants. However, according to experts, payers’ willingness to cover costly new therapies remains a work in progress.

The availability of more effective treatments with fewer side effects has brought about a cultural shift, said James F. Howard, Jr, MD. “The physician’s goal now is for the patient to be symptom free with grade 1 or less adverse events. And patients are demanding freedom from all the side effects that our usual course of immune therapy produces.” Dr. Howard is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill.

James F. Howard, Jr., MD, is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill in Chapel Hill, NC.
courtesy University of North Carolina
Dr. James F. Howard, Jr

The shift has been long in coming. Although myasthenia gravis was identified in the mid-1600s, it took more than 340 years to get the first drug approved specifically for the disorder.

Worldwide prevalence estimates vary widely, from less than 200,000 to 700,000 cases.1,2 Pathophysiologically, myasthenia gravis stems from autoimmune destruction of neuromuscular junctions (NMJs), which transmit motor neuron impulses to muscle fibers.1 Symptoms include variable skeletal muscle weakness that can range from mild and transient to life-threatening.

In approximately 80% of cases, autoimmune antibodies target the postsynaptic acetylcholine receptor (AChR). Additional autoimmune targets mainly include muscle-specific kinase (MuSK) and lipoprotein receptor-related protein 4 (LRP4). However, around 10% of patients are seronegative, lacking autoantibodies detectable through conventional radioimmunoassays. Clinical disease does not always correspond with circulating antibody levels, and pathogenesis may require cooperation between multiple autoantibodies attacking the same target.3 Around 10% of MG cases are associated with thymomas.

Among myasthenia gravis treatments, immunosuppressants typically take 4-10 months to begin working and 18-36 months for maximum benefit. “Our new targeted therapies work within 1-2 weeks, with maximum improvement occurring somewhere between 8 and 12 weeks,” Dr. Howard said. Quick onset makes these drugs well suited for primary therapy in recalcitrant myasthenia gravis or as bridges to standard immunotherapy. Targeted drugs also appear to provide effective rescue therapy, although head-to-head studies are needed.
 

Complement Inhibition

In AChR antibody–positive myasthenia gravis, autoantibody binding with the postsynaptic AChR receptor activates complement to attack postsynaptic neuronal membrane. Complement inhibitors approved to date block activation of the terminal complement protein C5.

Henry J. Kaminski, MD, is is Meta A. Neumann Professor of Neurology at George Washington University in Washington, DC.
courtesy George Washington University
Dr. Henry J. Kaminski

For many patients, complement inhibitors deliver dramatic results. Henry J. Kaminski, MD, said that the first patient for whom he prescribed a complement inhibitor outside a clinical trial went from being miserable to traveling internationally within a month. Dr. Kaminski is Meta A. Neumann Professor of Neurology at George Washington University, Washington, DC. 

Eculizumab (Soliris, Alexion), earned Food and Drug Administration (FDA) approval for myasthenia gravis in 2017. Week 26 results in the phase 3 REGAIN trial showed no significant difference in Myasthenia Gravis–Activities of Daily Living (MG-ADL) scores between treatment and placebo. However, said Dr. Howard, primary investigator on the study, the negative result was a statistical aberration stemming from the FDA’s requirement to use worst-rank analysis rather than absolute change scores. What got eculizumab approved were highly positive results in the overwhelming majority of secondary endpoints.4 Subsequently, the FDA had the manufacturer rewrite the package insert using common statistical methods, which yielded positive primary results.

Ravulizumab (Ultomiris, Alexion), approved for myasthenia gravis in 2022, reduces eculizumab’s twice-monthly intravenous dosing to every 2 months (after loading doses), with very similar efficacy. The newest complement inhibitor, zilucoplan (Zilbrysq, UCB), administered once daily subcutaneously, earned FDA approval in 2023. Daily subcutaneous dosing provides patient convenience, said Dr. Howard. Because the body does not clear this small molecule as it would a full-size antibody, it is the only complement inhibitor that can be combined with a fragment crystallizable neonatal receptor (FcRn) inhibitor.
 

 

 

FcRn Inhibition

The FcRn exists on the surface and intracellular vesicles of many cells, including B cells, but not T cells.5FcRn inhibitors block binding of circulating IgG antibodies to the FcRn, preventing their normal recycling, significantly reducing circulating antibodies within days of treatment.

Efgartigimod (Vyvgart, Argenx), earned FDA approval in intravenous form in 2021, followed by a subcutaneous formulation that includes hyaluronidase (Vyvgart Hytrulo) in 2023. Rozanolixizumab (Rystiggo, UCB) earned FDA approval for both AChR antibody–positive and MuSK antibody–positive myasthenia gravis in 2023.

Along with rapid response, said Dr. Howard, complement inhibitors and FcRn inhibitors offer a “hugely improved” side-effect profile. In phase 3 research, the most common side effects for both classes included headache, nausea, and diarrhea.4,6,7 Because complement inhibitors increase the risk of Neisseria infection, users require immunization against meningococcal infection (or concurrent antibiotic prophylaxis) while on complement inhibitors.
 

Insurance Issues

With many clinicians wondering which targeted therapy to choose for a particular patient, said Dr. Howard and Dr. Kaminski, the main obstacle to wider use of these treatments is payer attitudes and practices. “While many of us would like to see these drugs used earlier in the course of disease,” Dr. Howard explained, “there are numerous restrictions placed on the physician and the patient by whatever insurance the individual has.”

Dr. Kaminski said: “There’s a lot of variability among insurance companies regarding what is expected in terms of getting approval for a certain medication. It frustrates me, thinking this patient may do well with a complement inhibitor or an FcRn inhibitor, but it takes weeks to get them approved.”

Some of his patients have been approved for, and flourished on, complement inhibitors and FcRn inhibitors, he added, and then denied a second round of treatment. Dr. Kaminski said he does not know why these patients were denied, and every time he requests reevaluation, the decision is reversed. “That’s a significant time frame for me and my staff to manage.”

When asked what can be done to address high drug prices, Dr. Howard replied, “I have no idea. I’m not an advocate of high drug prices. But I don’t think people realize the cost of doing clinical trials, which is hundreds of millions of dollars, particularly in rare diseases.”

Presently, Dr. Howard said, FcRn inhibitors are used more frequently than complement inhibitors solely because of cost. Zilucoplan will be priced below existing complement inhibitors, although it is too soon to compare its price with those of FcRn inhibitors.

When eculizumab debuted, said Dr. Howard, it cost nearly $750,000 annually. “But if you look at the number of patients treated, the cost of the drug over this population is probably less than the cost for using a cholesterol-lowering agent to treat hyperlipidemia.”

An Institute for Clinical and Economic Review (ICER) report stated that eculizumab and efgartigimod should both cost less than $20,000 annually to meet commonly used cost-effectiveness thresholds.8 However, Dr. Howard said ICER used models based on common diseases and ignored the economic impact of patients’ losing fewer workdays and avoiding long-term immunosuppressant side effects such as diabetes and osteoporosis and related treatment costs. “We’ve got to start looking at total societal cost,” he said.

 

 

Leapfrogging Ahead

Not all the new drugs work in every indicated patient, Dr. Howard said. For example, up to 30% of patients do not respond to complement inhibitors. “We don’t understand why. It’s as if we have leapfrogged way ahead in terms of therapeutics, and now we have to go back and answer all the questions – the who, what, where, and why of an individual drug and its response in folks.”

In this climate, said Dr. Kaminski, heavy direct-to-consumer advertising of newer myasthenia gravis therapies creates complications. “My patients are highly excited to see, ‘that’s my disease being advertised on Jeopardy.’ ” Many patients are frustrated with the general lack of awareness regarding myasthenia gravis, he added. “But then I’ve had patients who clearly would never qualify for a certain medication getting mailings to their homes.”

Dr. Howard countered that broader awareness of myasthenia gravis can only help. “There’s increasing recognition of the disease, not only by patients, but to some extent, by the treating clinician. Patients are coming to our offices and saying, ‘am I a candidate for this new drug?’ It’s the responsibility of the clinician to decide.”

Individual physicians’ practice patterns vary greatly, said Dr. Kaminski, and very little quantitative data exist here. But based on personal communications, academic-center neurologists tend to use targeted treatments on patients who have failed conventional treatments.

Conversely, Dr. Howard said that, because community physicians rarely see myasthenia gravis, and targeted treatments remain relatively new, many of these providers rely on prednisone, azathioprine, and mycophenolate mofetil.
 

B-Cell Blockers in Development

Overall, said Dr. Howard, the field of myasthenia gravis treatment development is “very rich. And pharma’s interest in myasthenia has taken off like a rocket. It’s exceptionally gratifying to those of us who take care of these patients whose life is miserable” because of adverse effects and/or nonresponse to current drugs.

“In myasthenia,” added Dr. Kaminski, “we know that T cells are promoting the activity of these auto-reactive B cells.” Many drugs currently in phase 2 or 3 development aim to eliminate B cells or signaling between T and B cells, he said. “That’s where most of the drug development is.”

Leading candidates include telitacicept (Tai’ai, RemeGen), which is both a B-lymphocyte stimulator and a proliferation-inducing ligand. A phase 3 trial (NCT05737160) is ongoing, with primary completion expected in late 2026. A second phase 3 trial (NCT06456580) recently began enrolling. Dr. Howard said that, although early results warranted phase 3 analysis, telitacicept’s phase 2 trial was open label and lacked a placebo group.9 The latter is a critical concern because placebo response rates in myasthenia gravis trials average 35%-40%.

Combined with standard care, the FcRn inhibitor nipocalimab (Johnson & Johnson) enabled patients with AChR, MuSK, and/or LRP4 autoantibodies to improve by 4.70 points on the MG-ADL vs 3.25 points for placebo (P = .002) over 24 weeks in phase 3.10All FcRn inhibitors in development can broadly reduce autoantibody levels, said Dr. Howard. “But what role they will play in myasthenia gravis when they’re several years behind leaders in the field in terms of capturing market remains to be seen.”

Additionally, batoclimab (Immunovant/Harbour BioMed) showed positive topline results in phase 3, and an elevated rate of hypercholesterolemia in treated patients that was transient and consistent with previous research.11 Subsequent to efgartigimod, Dr. Howard said, FcRn inhibitors are full-size antibodies. “I believe that contributes to the adverse events that we see. Efgartigimod is a small FcRn fragment. That’s why it’s a cleaner drug, if you will.”

FcRn inhibitors require periodic retreatment. For example, said Dr. Howard, the ADAPT phase 3 trial of efgartigimod, on which he was lead investigator, employed a cyclic dosing schedule – 4 weeks’ treatment, then observation until patients needed retreatment — because patients demanded it.12 In clinical practice, some patients have gone more than 25 weeks before needing retreatment. One of his patients went beyond 40 weeks. “Others only get around 6-9 weeks. So patient choice again enters the decision-making process.”

Rituximab targets the CD20 protein on B cells nonspecifically, producing general immunosuppression. “That’s problematic in producing significant immunosuppression,” said Dr. Kaminski. Nevertheless, he said, rituximab is very effective for most patients with MuSK-specific MG, and its application to this indication has revealed differences between the MuSK subtype and AChR antibody–positive myasthenia. Specifically, MuSK antibody–positive patients have short-lived plasmablasts, which rituximab eliminates.13

Conversely, said Dr. Kaminski, patients with AChR antibody-positive myasthenia, especially long-term, likely have long-lived plasmablasts producing antibodies. This fact, and these patients’ lack of CD20, likely explain their poor response to rituximab.

A phase 3 trial (NCT04524273) of the CD19 blocker inebilizumab (Uplinza, Amgen) reached primary completion in May. Dr. Howard said that if topline results (unreleased at press time) prove positive, inebilizumab could replace rituximab in MG — provided payers do not reject inebilizumab because of cost.

 

 


Packed Early-Development Pipeline

Regarding early-stage projects, said Dr. Howard, the pipeline is packed with compounds that target various aspects of the immune system. “The real question with those is, what’s going to be the side effect profile? All of the trials are very early. We need bigger trials with much longer observation for safety, durability, and degree of efficacy.”

The next potential B cell–targeting game changer, he said, is chimeric antigen receptor (CAR) T cell–based therapy. In a phase 2b trial of Descartes-08 (Cartesian Therapeutics), 71% of treated patients experienced clinically meaningful improvement in MG Composite score at 3 months vs 25% for placebo.14

In early clinical trials, said Dr. Howard, patients treated with Descartes-08 — which uses autologous mRNA to target B-cell maturation antigen — have shown “exceptional improvement” lasting 20 or more months. Because the drug is not ingrained permanently into the genome, Descartes-08 avoids potentially severe side effects of DNA-targeting CAR T candidates. Dr. Howard hopes a phase 3 trial will commence around January 2025.

The tolerance approach exemplified by CNP-106 (COUR Pharmaceuticals) and a myasthenia gravis tolerogen (Toleranzia) seeks to prevent the immune system from recognizing and reacting to the NMJ abnormalities that produce myasthenia gravis, potentially providing a cure. “We look forward to those trials as they come online in the next 1-2 years,” said Dr. Howard.
 

Unmet Needs

Historically, neurologists believed that all myasthenia gravis symptoms stemmed from muscle fatigue — the more active the muscle, the weaker it gets. However, said Dr. Kaminski, some patients might lack measurable weakness but still complain of fatigue.

Elevated levels of cytokines such as interleukin (IL)–6 or IL-17 also can produce fatigue, he noted. “With the drugs we’re using, certainly the new ones, we’re not specifically targeting this fatigue phenomenon, which has been studied in a very limited fashion.”

In the RAISE-XT zilucoplan trial, participants experienced significant improvement in fatigue scores for up to 60 weeks.15 Although zilucoplan does not address fatigue directly, said Dr. Howard, improving myasthenia gravis overall helps reduce fatigue.

The Myasthenia Gravis Symptoms Patient Reported Outcome (MG Symptoms PRO), which Dr. Kaminski helped develop, includes questions designed to distinguish muscular fatigue from overall physical fatigue.16 “I’m very interested in some of the information that’s coming out on long COVID and its effect on muscle,” Dr. Kaminski added. “We might be able to learn from there that there’s still some pathology going on beyond the neuromuscular junction.”

What the field desperately needs, said Dr. Howard, are biomarkers to identify which patients will and will not respond to certain therapeutics. “We’re not there yet.” Such biomarkers are at least 3-7 years from becoming clinical reality.

Promising antibody-independent serum markers include circulating microRNAs. For example, miRNA-150-5p and miRNA-21-5p are elevated in generalized AChR-positive myasthenia gravis and early-onset myasthenia gravis (occurring before age 50) and decline after immunosuppression and thymectomy.17

Among diagnostic modalities for patients with seronegative myasthenia gravis, said Dr. Kaminski, single-fiber EMG is the most sensitive, at approximately 95%. “It’s not perfect.” Moreover, he said, performing this test accurately requires a highly experienced expert, which many treatment centers lack.

Presently, added Dr. Kaminski, orbital MRI is neither specific nor sensitive enough to be clinically useful. “One needs to be careful with these specialized tests that are published from the best laboratory in the world that does the test, and does it repetitively.” As the search for effective myasthenia gravis biomarkers continues, avoiding false-positive results is as important as avoiding false negatives.

 

References

1. Bubuioc AM et al. J Med Life. 2021 Jan-Mar;14(1):7-16. doi: 10.25122/jml-2020-0145.

2. Deenen JC et al. J Neuromuscul Dis. 2015;2(1):73-85. doi: 10.3233/JND-140045.

3. Kaminski HJ et al. J Clin Invest. 2024 Jun 17;134(12):e179742. doi: 10.1172/JCI179742.

4. Howard JF Jr et al. Lancet Neurol. 2017 Dec;16(12):976-986. doi: 10.1016/S1474-4422(17)30369-1.

5. Huda R. Front Immunol. 2020 Feb 21:11:240. doi: 10.3389/fimmu.2020.00240.

6. Howard JF Jr et al. Lancet Neurol. 2023 May;22(5):395-406. doi: 10.1016/S1474-4422(23)00080-7.

7. Vu T et al. NEJM Evid. 2022 May;1(5):EVIDoa2100066. doi: 10.1056/EVIDoa2100066.

8. Tice JA et al. October 20, 2021. https://icer.org/assessment/myasthenia-gravis/.

9. Yin J et al. Eur J Neurol. 2024 Aug;31(8):e16322. doi: 10.1111/ene.16322.

10. Antozzi C et al. EAN 2024, Abstract EPR-116. https://www.neurology.org/doi/10.1212/WNL.0000000000203660.

11. Yan C et al. JAMA Neurol. 2024 Mar 4;81(4):336-345. doi: 10.1001/jamaneurol.2024.0044.

12. Howard JF Jr et al. Lancet Neurol. 2021 Jul;20(7):526-536. doi: 10.1016/S1474-4422(21)00159-9.

13. Stathopoulos P et al. JCI Insight. 2017 Sep 7;2(17):e94263. doi: 10.1172/jci.insight.94263.

14. Cartesian Therapeutics. Cartesian Therapeutics announces positive topline results from phase 2b trial of Descartes-08 in patients with myasthenia gravis. 2024 Jul 2. https://ir.cartesiantherapeutics.com/news-releases/news-release-details/cartesian-therapeutics-announces-positive-topline-results-phase.

15. Howard JF Jr et al. Ther Adv Neurol Disord. 2024 Apr 17:17:17562864241243186. doi: 10.1177/17562864241243186.

16. Cleanthous S et al. Orphanet J Rare Dis. 2021 Oct 30;16(1):457. doi: 10.1186/s13023-021-02064-0.

17. Sabre L et al. Front Immunol. 2020 Mar 4:11:213. doi: 10.3389/fimmu.2020.00213.

Publications
Topics
Sections

Unlike conventional immunosuppression, treatments approved since 2017 are giving patients with myasthenia gravis targeted options to better match their needs, desires, and tolerance. Used appropriately, newer treatments can provide dramatic results faster and more safely than broad immunosuppressants. However, according to experts, payers’ willingness to cover costly new therapies remains a work in progress.

The availability of more effective treatments with fewer side effects has brought about a cultural shift, said James F. Howard, Jr, MD. “The physician’s goal now is for the patient to be symptom free with grade 1 or less adverse events. And patients are demanding freedom from all the side effects that our usual course of immune therapy produces.” Dr. Howard is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill.

James F. Howard, Jr., MD, is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill in Chapel Hill, NC.
courtesy University of North Carolina
Dr. James F. Howard, Jr

The shift has been long in coming. Although myasthenia gravis was identified in the mid-1600s, it took more than 340 years to get the first drug approved specifically for the disorder.

Worldwide prevalence estimates vary widely, from less than 200,000 to 700,000 cases.1,2 Pathophysiologically, myasthenia gravis stems from autoimmune destruction of neuromuscular junctions (NMJs), which transmit motor neuron impulses to muscle fibers.1 Symptoms include variable skeletal muscle weakness that can range from mild and transient to life-threatening.

In approximately 80% of cases, autoimmune antibodies target the postsynaptic acetylcholine receptor (AChR). Additional autoimmune targets mainly include muscle-specific kinase (MuSK) and lipoprotein receptor-related protein 4 (LRP4). However, around 10% of patients are seronegative, lacking autoantibodies detectable through conventional radioimmunoassays. Clinical disease does not always correspond with circulating antibody levels, and pathogenesis may require cooperation between multiple autoantibodies attacking the same target.3 Around 10% of MG cases are associated with thymomas.

Among myasthenia gravis treatments, immunosuppressants typically take 4-10 months to begin working and 18-36 months for maximum benefit. “Our new targeted therapies work within 1-2 weeks, with maximum improvement occurring somewhere between 8 and 12 weeks,” Dr. Howard said. Quick onset makes these drugs well suited for primary therapy in recalcitrant myasthenia gravis or as bridges to standard immunotherapy. Targeted drugs also appear to provide effective rescue therapy, although head-to-head studies are needed.
 

Complement Inhibition

In AChR antibody–positive myasthenia gravis, autoantibody binding with the postsynaptic AChR receptor activates complement to attack postsynaptic neuronal membrane. Complement inhibitors approved to date block activation of the terminal complement protein C5.

Henry J. Kaminski, MD, is is Meta A. Neumann Professor of Neurology at George Washington University in Washington, DC.
courtesy George Washington University
Dr. Henry J. Kaminski

For many patients, complement inhibitors deliver dramatic results. Henry J. Kaminski, MD, said that the first patient for whom he prescribed a complement inhibitor outside a clinical trial went from being miserable to traveling internationally within a month. Dr. Kaminski is Meta A. Neumann Professor of Neurology at George Washington University, Washington, DC. 

Eculizumab (Soliris, Alexion), earned Food and Drug Administration (FDA) approval for myasthenia gravis in 2017. Week 26 results in the phase 3 REGAIN trial showed no significant difference in Myasthenia Gravis–Activities of Daily Living (MG-ADL) scores between treatment and placebo. However, said Dr. Howard, primary investigator on the study, the negative result was a statistical aberration stemming from the FDA’s requirement to use worst-rank analysis rather than absolute change scores. What got eculizumab approved were highly positive results in the overwhelming majority of secondary endpoints.4 Subsequently, the FDA had the manufacturer rewrite the package insert using common statistical methods, which yielded positive primary results.

Ravulizumab (Ultomiris, Alexion), approved for myasthenia gravis in 2022, reduces eculizumab’s twice-monthly intravenous dosing to every 2 months (after loading doses), with very similar efficacy. The newest complement inhibitor, zilucoplan (Zilbrysq, UCB), administered once daily subcutaneously, earned FDA approval in 2023. Daily subcutaneous dosing provides patient convenience, said Dr. Howard. Because the body does not clear this small molecule as it would a full-size antibody, it is the only complement inhibitor that can be combined with a fragment crystallizable neonatal receptor (FcRn) inhibitor.
 

 

 

FcRn Inhibition

The FcRn exists on the surface and intracellular vesicles of many cells, including B cells, but not T cells.5FcRn inhibitors block binding of circulating IgG antibodies to the FcRn, preventing their normal recycling, significantly reducing circulating antibodies within days of treatment.

Efgartigimod (Vyvgart, Argenx), earned FDA approval in intravenous form in 2021, followed by a subcutaneous formulation that includes hyaluronidase (Vyvgart Hytrulo) in 2023. Rozanolixizumab (Rystiggo, UCB) earned FDA approval for both AChR antibody–positive and MuSK antibody–positive myasthenia gravis in 2023.

Along with rapid response, said Dr. Howard, complement inhibitors and FcRn inhibitors offer a “hugely improved” side-effect profile. In phase 3 research, the most common side effects for both classes included headache, nausea, and diarrhea.4,6,7 Because complement inhibitors increase the risk of Neisseria infection, users require immunization against meningococcal infection (or concurrent antibiotic prophylaxis) while on complement inhibitors.
 

Insurance Issues

With many clinicians wondering which targeted therapy to choose for a particular patient, said Dr. Howard and Dr. Kaminski, the main obstacle to wider use of these treatments is payer attitudes and practices. “While many of us would like to see these drugs used earlier in the course of disease,” Dr. Howard explained, “there are numerous restrictions placed on the physician and the patient by whatever insurance the individual has.”

Dr. Kaminski said: “There’s a lot of variability among insurance companies regarding what is expected in terms of getting approval for a certain medication. It frustrates me, thinking this patient may do well with a complement inhibitor or an FcRn inhibitor, but it takes weeks to get them approved.”

Some of his patients have been approved for, and flourished on, complement inhibitors and FcRn inhibitors, he added, and then denied a second round of treatment. Dr. Kaminski said he does not know why these patients were denied, and every time he requests reevaluation, the decision is reversed. “That’s a significant time frame for me and my staff to manage.”

When asked what can be done to address high drug prices, Dr. Howard replied, “I have no idea. I’m not an advocate of high drug prices. But I don’t think people realize the cost of doing clinical trials, which is hundreds of millions of dollars, particularly in rare diseases.”

Presently, Dr. Howard said, FcRn inhibitors are used more frequently than complement inhibitors solely because of cost. Zilucoplan will be priced below existing complement inhibitors, although it is too soon to compare its price with those of FcRn inhibitors.

When eculizumab debuted, said Dr. Howard, it cost nearly $750,000 annually. “But if you look at the number of patients treated, the cost of the drug over this population is probably less than the cost for using a cholesterol-lowering agent to treat hyperlipidemia.”

An Institute for Clinical and Economic Review (ICER) report stated that eculizumab and efgartigimod should both cost less than $20,000 annually to meet commonly used cost-effectiveness thresholds.8 However, Dr. Howard said ICER used models based on common diseases and ignored the economic impact of patients’ losing fewer workdays and avoiding long-term immunosuppressant side effects such as diabetes and osteoporosis and related treatment costs. “We’ve got to start looking at total societal cost,” he said.

 

 

Leapfrogging Ahead

Not all the new drugs work in every indicated patient, Dr. Howard said. For example, up to 30% of patients do not respond to complement inhibitors. “We don’t understand why. It’s as if we have leapfrogged way ahead in terms of therapeutics, and now we have to go back and answer all the questions – the who, what, where, and why of an individual drug and its response in folks.”

In this climate, said Dr. Kaminski, heavy direct-to-consumer advertising of newer myasthenia gravis therapies creates complications. “My patients are highly excited to see, ‘that’s my disease being advertised on Jeopardy.’ ” Many patients are frustrated with the general lack of awareness regarding myasthenia gravis, he added. “But then I’ve had patients who clearly would never qualify for a certain medication getting mailings to their homes.”

Dr. Howard countered that broader awareness of myasthenia gravis can only help. “There’s increasing recognition of the disease, not only by patients, but to some extent, by the treating clinician. Patients are coming to our offices and saying, ‘am I a candidate for this new drug?’ It’s the responsibility of the clinician to decide.”

Individual physicians’ practice patterns vary greatly, said Dr. Kaminski, and very little quantitative data exist here. But based on personal communications, academic-center neurologists tend to use targeted treatments on patients who have failed conventional treatments.

Conversely, Dr. Howard said that, because community physicians rarely see myasthenia gravis, and targeted treatments remain relatively new, many of these providers rely on prednisone, azathioprine, and mycophenolate mofetil.
 

B-Cell Blockers in Development

Overall, said Dr. Howard, the field of myasthenia gravis treatment development is “very rich. And pharma’s interest in myasthenia has taken off like a rocket. It’s exceptionally gratifying to those of us who take care of these patients whose life is miserable” because of adverse effects and/or nonresponse to current drugs.

“In myasthenia,” added Dr. Kaminski, “we know that T cells are promoting the activity of these auto-reactive B cells.” Many drugs currently in phase 2 or 3 development aim to eliminate B cells or signaling between T and B cells, he said. “That’s where most of the drug development is.”

Leading candidates include telitacicept (Tai’ai, RemeGen), which is both a B-lymphocyte stimulator and a proliferation-inducing ligand. A phase 3 trial (NCT05737160) is ongoing, with primary completion expected in late 2026. A second phase 3 trial (NCT06456580) recently began enrolling. Dr. Howard said that, although early results warranted phase 3 analysis, telitacicept’s phase 2 trial was open label and lacked a placebo group.9 The latter is a critical concern because placebo response rates in myasthenia gravis trials average 35%-40%.

Combined with standard care, the FcRn inhibitor nipocalimab (Johnson & Johnson) enabled patients with AChR, MuSK, and/or LRP4 autoantibodies to improve by 4.70 points on the MG-ADL vs 3.25 points for placebo (P = .002) over 24 weeks in phase 3.10All FcRn inhibitors in development can broadly reduce autoantibody levels, said Dr. Howard. “But what role they will play in myasthenia gravis when they’re several years behind leaders in the field in terms of capturing market remains to be seen.”

Additionally, batoclimab (Immunovant/Harbour BioMed) showed positive topline results in phase 3, and an elevated rate of hypercholesterolemia in treated patients that was transient and consistent with previous research.11 Subsequent to efgartigimod, Dr. Howard said, FcRn inhibitors are full-size antibodies. “I believe that contributes to the adverse events that we see. Efgartigimod is a small FcRn fragment. That’s why it’s a cleaner drug, if you will.”

FcRn inhibitors require periodic retreatment. For example, said Dr. Howard, the ADAPT phase 3 trial of efgartigimod, on which he was lead investigator, employed a cyclic dosing schedule – 4 weeks’ treatment, then observation until patients needed retreatment — because patients demanded it.12 In clinical practice, some patients have gone more than 25 weeks before needing retreatment. One of his patients went beyond 40 weeks. “Others only get around 6-9 weeks. So patient choice again enters the decision-making process.”

Rituximab targets the CD20 protein on B cells nonspecifically, producing general immunosuppression. “That’s problematic in producing significant immunosuppression,” said Dr. Kaminski. Nevertheless, he said, rituximab is very effective for most patients with MuSK-specific MG, and its application to this indication has revealed differences between the MuSK subtype and AChR antibody–positive myasthenia. Specifically, MuSK antibody–positive patients have short-lived plasmablasts, which rituximab eliminates.13

Conversely, said Dr. Kaminski, patients with AChR antibody-positive myasthenia, especially long-term, likely have long-lived plasmablasts producing antibodies. This fact, and these patients’ lack of CD20, likely explain their poor response to rituximab.

A phase 3 trial (NCT04524273) of the CD19 blocker inebilizumab (Uplinza, Amgen) reached primary completion in May. Dr. Howard said that if topline results (unreleased at press time) prove positive, inebilizumab could replace rituximab in MG — provided payers do not reject inebilizumab because of cost.

 

 


Packed Early-Development Pipeline

Regarding early-stage projects, said Dr. Howard, the pipeline is packed with compounds that target various aspects of the immune system. “The real question with those is, what’s going to be the side effect profile? All of the trials are very early. We need bigger trials with much longer observation for safety, durability, and degree of efficacy.”

The next potential B cell–targeting game changer, he said, is chimeric antigen receptor (CAR) T cell–based therapy. In a phase 2b trial of Descartes-08 (Cartesian Therapeutics), 71% of treated patients experienced clinically meaningful improvement in MG Composite score at 3 months vs 25% for placebo.14

In early clinical trials, said Dr. Howard, patients treated with Descartes-08 — which uses autologous mRNA to target B-cell maturation antigen — have shown “exceptional improvement” lasting 20 or more months. Because the drug is not ingrained permanently into the genome, Descartes-08 avoids potentially severe side effects of DNA-targeting CAR T candidates. Dr. Howard hopes a phase 3 trial will commence around January 2025.

The tolerance approach exemplified by CNP-106 (COUR Pharmaceuticals) and a myasthenia gravis tolerogen (Toleranzia) seeks to prevent the immune system from recognizing and reacting to the NMJ abnormalities that produce myasthenia gravis, potentially providing a cure. “We look forward to those trials as they come online in the next 1-2 years,” said Dr. Howard.
 

Unmet Needs

Historically, neurologists believed that all myasthenia gravis symptoms stemmed from muscle fatigue — the more active the muscle, the weaker it gets. However, said Dr. Kaminski, some patients might lack measurable weakness but still complain of fatigue.

Elevated levels of cytokines such as interleukin (IL)–6 or IL-17 also can produce fatigue, he noted. “With the drugs we’re using, certainly the new ones, we’re not specifically targeting this fatigue phenomenon, which has been studied in a very limited fashion.”

In the RAISE-XT zilucoplan trial, participants experienced significant improvement in fatigue scores for up to 60 weeks.15 Although zilucoplan does not address fatigue directly, said Dr. Howard, improving myasthenia gravis overall helps reduce fatigue.

The Myasthenia Gravis Symptoms Patient Reported Outcome (MG Symptoms PRO), which Dr. Kaminski helped develop, includes questions designed to distinguish muscular fatigue from overall physical fatigue.16 “I’m very interested in some of the information that’s coming out on long COVID and its effect on muscle,” Dr. Kaminski added. “We might be able to learn from there that there’s still some pathology going on beyond the neuromuscular junction.”

What the field desperately needs, said Dr. Howard, are biomarkers to identify which patients will and will not respond to certain therapeutics. “We’re not there yet.” Such biomarkers are at least 3-7 years from becoming clinical reality.

Promising antibody-independent serum markers include circulating microRNAs. For example, miRNA-150-5p and miRNA-21-5p are elevated in generalized AChR-positive myasthenia gravis and early-onset myasthenia gravis (occurring before age 50) and decline after immunosuppression and thymectomy.17

Among diagnostic modalities for patients with seronegative myasthenia gravis, said Dr. Kaminski, single-fiber EMG is the most sensitive, at approximately 95%. “It’s not perfect.” Moreover, he said, performing this test accurately requires a highly experienced expert, which many treatment centers lack.

Presently, added Dr. Kaminski, orbital MRI is neither specific nor sensitive enough to be clinically useful. “One needs to be careful with these specialized tests that are published from the best laboratory in the world that does the test, and does it repetitively.” As the search for effective myasthenia gravis biomarkers continues, avoiding false-positive results is as important as avoiding false negatives.

 

References

1. Bubuioc AM et al. J Med Life. 2021 Jan-Mar;14(1):7-16. doi: 10.25122/jml-2020-0145.

2. Deenen JC et al. J Neuromuscul Dis. 2015;2(1):73-85. doi: 10.3233/JND-140045.

3. Kaminski HJ et al. J Clin Invest. 2024 Jun 17;134(12):e179742. doi: 10.1172/JCI179742.

4. Howard JF Jr et al. Lancet Neurol. 2017 Dec;16(12):976-986. doi: 10.1016/S1474-4422(17)30369-1.

5. Huda R. Front Immunol. 2020 Feb 21:11:240. doi: 10.3389/fimmu.2020.00240.

6. Howard JF Jr et al. Lancet Neurol. 2023 May;22(5):395-406. doi: 10.1016/S1474-4422(23)00080-7.

7. Vu T et al. NEJM Evid. 2022 May;1(5):EVIDoa2100066. doi: 10.1056/EVIDoa2100066.

8. Tice JA et al. October 20, 2021. https://icer.org/assessment/myasthenia-gravis/.

9. Yin J et al. Eur J Neurol. 2024 Aug;31(8):e16322. doi: 10.1111/ene.16322.

10. Antozzi C et al. EAN 2024, Abstract EPR-116. https://www.neurology.org/doi/10.1212/WNL.0000000000203660.

11. Yan C et al. JAMA Neurol. 2024 Mar 4;81(4):336-345. doi: 10.1001/jamaneurol.2024.0044.

12. Howard JF Jr et al. Lancet Neurol. 2021 Jul;20(7):526-536. doi: 10.1016/S1474-4422(21)00159-9.

13. Stathopoulos P et al. JCI Insight. 2017 Sep 7;2(17):e94263. doi: 10.1172/jci.insight.94263.

14. Cartesian Therapeutics. Cartesian Therapeutics announces positive topline results from phase 2b trial of Descartes-08 in patients with myasthenia gravis. 2024 Jul 2. https://ir.cartesiantherapeutics.com/news-releases/news-release-details/cartesian-therapeutics-announces-positive-topline-results-phase.

15. Howard JF Jr et al. Ther Adv Neurol Disord. 2024 Apr 17:17:17562864241243186. doi: 10.1177/17562864241243186.

16. Cleanthous S et al. Orphanet J Rare Dis. 2021 Oct 30;16(1):457. doi: 10.1186/s13023-021-02064-0.

17. Sabre L et al. Front Immunol. 2020 Mar 4:11:213. doi: 10.3389/fimmu.2020.00213.

Unlike conventional immunosuppression, treatments approved since 2017 are giving patients with myasthenia gravis targeted options to better match their needs, desires, and tolerance. Used appropriately, newer treatments can provide dramatic results faster and more safely than broad immunosuppressants. However, according to experts, payers’ willingness to cover costly new therapies remains a work in progress.

The availability of more effective treatments with fewer side effects has brought about a cultural shift, said James F. Howard, Jr, MD. “The physician’s goal now is for the patient to be symptom free with grade 1 or less adverse events. And patients are demanding freedom from all the side effects that our usual course of immune therapy produces.” Dr. Howard is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill.

James F. Howard, Jr., MD, is professor of neurology, medicine and allied health and director of the Myasthenia Gravis Clinical Trials and Translational Research Program at the University of North Carolina at Chapel Hill in Chapel Hill, NC.
courtesy University of North Carolina
Dr. James F. Howard, Jr

The shift has been long in coming. Although myasthenia gravis was identified in the mid-1600s, it took more than 340 years to get the first drug approved specifically for the disorder.

Worldwide prevalence estimates vary widely, from less than 200,000 to 700,000 cases.1,2 Pathophysiologically, myasthenia gravis stems from autoimmune destruction of neuromuscular junctions (NMJs), which transmit motor neuron impulses to muscle fibers.1 Symptoms include variable skeletal muscle weakness that can range from mild and transient to life-threatening.

In approximately 80% of cases, autoimmune antibodies target the postsynaptic acetylcholine receptor (AChR). Additional autoimmune targets mainly include muscle-specific kinase (MuSK) and lipoprotein receptor-related protein 4 (LRP4). However, around 10% of patients are seronegative, lacking autoantibodies detectable through conventional radioimmunoassays. Clinical disease does not always correspond with circulating antibody levels, and pathogenesis may require cooperation between multiple autoantibodies attacking the same target.3 Around 10% of MG cases are associated with thymomas.

Among myasthenia gravis treatments, immunosuppressants typically take 4-10 months to begin working and 18-36 months for maximum benefit. “Our new targeted therapies work within 1-2 weeks, with maximum improvement occurring somewhere between 8 and 12 weeks,” Dr. Howard said. Quick onset makes these drugs well suited for primary therapy in recalcitrant myasthenia gravis or as bridges to standard immunotherapy. Targeted drugs also appear to provide effective rescue therapy, although head-to-head studies are needed.
 

Complement Inhibition

In AChR antibody–positive myasthenia gravis, autoantibody binding with the postsynaptic AChR receptor activates complement to attack postsynaptic neuronal membrane. Complement inhibitors approved to date block activation of the terminal complement protein C5.

Henry J. Kaminski, MD, is is Meta A. Neumann Professor of Neurology at George Washington University in Washington, DC.
courtesy George Washington University
Dr. Henry J. Kaminski

For many patients, complement inhibitors deliver dramatic results. Henry J. Kaminski, MD, said that the first patient for whom he prescribed a complement inhibitor outside a clinical trial went from being miserable to traveling internationally within a month. Dr. Kaminski is Meta A. Neumann Professor of Neurology at George Washington University, Washington, DC. 

Eculizumab (Soliris, Alexion), earned Food and Drug Administration (FDA) approval for myasthenia gravis in 2017. Week 26 results in the phase 3 REGAIN trial showed no significant difference in Myasthenia Gravis–Activities of Daily Living (MG-ADL) scores between treatment and placebo. However, said Dr. Howard, primary investigator on the study, the negative result was a statistical aberration stemming from the FDA’s requirement to use worst-rank analysis rather than absolute change scores. What got eculizumab approved were highly positive results in the overwhelming majority of secondary endpoints.4 Subsequently, the FDA had the manufacturer rewrite the package insert using common statistical methods, which yielded positive primary results.

Ravulizumab (Ultomiris, Alexion), approved for myasthenia gravis in 2022, reduces eculizumab’s twice-monthly intravenous dosing to every 2 months (after loading doses), with very similar efficacy. The newest complement inhibitor, zilucoplan (Zilbrysq, UCB), administered once daily subcutaneously, earned FDA approval in 2023. Daily subcutaneous dosing provides patient convenience, said Dr. Howard. Because the body does not clear this small molecule as it would a full-size antibody, it is the only complement inhibitor that can be combined with a fragment crystallizable neonatal receptor (FcRn) inhibitor.
 

 

 

FcRn Inhibition

The FcRn exists on the surface and intracellular vesicles of many cells, including B cells, but not T cells.5FcRn inhibitors block binding of circulating IgG antibodies to the FcRn, preventing their normal recycling, significantly reducing circulating antibodies within days of treatment.

Efgartigimod (Vyvgart, Argenx), earned FDA approval in intravenous form in 2021, followed by a subcutaneous formulation that includes hyaluronidase (Vyvgart Hytrulo) in 2023. Rozanolixizumab (Rystiggo, UCB) earned FDA approval for both AChR antibody–positive and MuSK antibody–positive myasthenia gravis in 2023.

Along with rapid response, said Dr. Howard, complement inhibitors and FcRn inhibitors offer a “hugely improved” side-effect profile. In phase 3 research, the most common side effects for both classes included headache, nausea, and diarrhea.4,6,7 Because complement inhibitors increase the risk of Neisseria infection, users require immunization against meningococcal infection (or concurrent antibiotic prophylaxis) while on complement inhibitors.
 

Insurance Issues

With many clinicians wondering which targeted therapy to choose for a particular patient, said Dr. Howard and Dr. Kaminski, the main obstacle to wider use of these treatments is payer attitudes and practices. “While many of us would like to see these drugs used earlier in the course of disease,” Dr. Howard explained, “there are numerous restrictions placed on the physician and the patient by whatever insurance the individual has.”

Dr. Kaminski said: “There’s a lot of variability among insurance companies regarding what is expected in terms of getting approval for a certain medication. It frustrates me, thinking this patient may do well with a complement inhibitor or an FcRn inhibitor, but it takes weeks to get them approved.”

Some of his patients have been approved for, and flourished on, complement inhibitors and FcRn inhibitors, he added, and then denied a second round of treatment. Dr. Kaminski said he does not know why these patients were denied, and every time he requests reevaluation, the decision is reversed. “That’s a significant time frame for me and my staff to manage.”

When asked what can be done to address high drug prices, Dr. Howard replied, “I have no idea. I’m not an advocate of high drug prices. But I don’t think people realize the cost of doing clinical trials, which is hundreds of millions of dollars, particularly in rare diseases.”

Presently, Dr. Howard said, FcRn inhibitors are used more frequently than complement inhibitors solely because of cost. Zilucoplan will be priced below existing complement inhibitors, although it is too soon to compare its price with those of FcRn inhibitors.

When eculizumab debuted, said Dr. Howard, it cost nearly $750,000 annually. “But if you look at the number of patients treated, the cost of the drug over this population is probably less than the cost for using a cholesterol-lowering agent to treat hyperlipidemia.”

An Institute for Clinical and Economic Review (ICER) report stated that eculizumab and efgartigimod should both cost less than $20,000 annually to meet commonly used cost-effectiveness thresholds.8 However, Dr. Howard said ICER used models based on common diseases and ignored the economic impact of patients’ losing fewer workdays and avoiding long-term immunosuppressant side effects such as diabetes and osteoporosis and related treatment costs. “We’ve got to start looking at total societal cost,” he said.

 

 

Leapfrogging Ahead

Not all the new drugs work in every indicated patient, Dr. Howard said. For example, up to 30% of patients do not respond to complement inhibitors. “We don’t understand why. It’s as if we have leapfrogged way ahead in terms of therapeutics, and now we have to go back and answer all the questions – the who, what, where, and why of an individual drug and its response in folks.”

In this climate, said Dr. Kaminski, heavy direct-to-consumer advertising of newer myasthenia gravis therapies creates complications. “My patients are highly excited to see, ‘that’s my disease being advertised on Jeopardy.’ ” Many patients are frustrated with the general lack of awareness regarding myasthenia gravis, he added. “But then I’ve had patients who clearly would never qualify for a certain medication getting mailings to their homes.”

Dr. Howard countered that broader awareness of myasthenia gravis can only help. “There’s increasing recognition of the disease, not only by patients, but to some extent, by the treating clinician. Patients are coming to our offices and saying, ‘am I a candidate for this new drug?’ It’s the responsibility of the clinician to decide.”

Individual physicians’ practice patterns vary greatly, said Dr. Kaminski, and very little quantitative data exist here. But based on personal communications, academic-center neurologists tend to use targeted treatments on patients who have failed conventional treatments.

Conversely, Dr. Howard said that, because community physicians rarely see myasthenia gravis, and targeted treatments remain relatively new, many of these providers rely on prednisone, azathioprine, and mycophenolate mofetil.
 

B-Cell Blockers in Development

Overall, said Dr. Howard, the field of myasthenia gravis treatment development is “very rich. And pharma’s interest in myasthenia has taken off like a rocket. It’s exceptionally gratifying to those of us who take care of these patients whose life is miserable” because of adverse effects and/or nonresponse to current drugs.

“In myasthenia,” added Dr. Kaminski, “we know that T cells are promoting the activity of these auto-reactive B cells.” Many drugs currently in phase 2 or 3 development aim to eliminate B cells or signaling between T and B cells, he said. “That’s where most of the drug development is.”

Leading candidates include telitacicept (Tai’ai, RemeGen), which is both a B-lymphocyte stimulator and a proliferation-inducing ligand. A phase 3 trial (NCT05737160) is ongoing, with primary completion expected in late 2026. A second phase 3 trial (NCT06456580) recently began enrolling. Dr. Howard said that, although early results warranted phase 3 analysis, telitacicept’s phase 2 trial was open label and lacked a placebo group.9 The latter is a critical concern because placebo response rates in myasthenia gravis trials average 35%-40%.

Combined with standard care, the FcRn inhibitor nipocalimab (Johnson & Johnson) enabled patients with AChR, MuSK, and/or LRP4 autoantibodies to improve by 4.70 points on the MG-ADL vs 3.25 points for placebo (P = .002) over 24 weeks in phase 3.10All FcRn inhibitors in development can broadly reduce autoantibody levels, said Dr. Howard. “But what role they will play in myasthenia gravis when they’re several years behind leaders in the field in terms of capturing market remains to be seen.”

Additionally, batoclimab (Immunovant/Harbour BioMed) showed positive topline results in phase 3, and an elevated rate of hypercholesterolemia in treated patients that was transient and consistent with previous research.11 Subsequent to efgartigimod, Dr. Howard said, FcRn inhibitors are full-size antibodies. “I believe that contributes to the adverse events that we see. Efgartigimod is a small FcRn fragment. That’s why it’s a cleaner drug, if you will.”

FcRn inhibitors require periodic retreatment. For example, said Dr. Howard, the ADAPT phase 3 trial of efgartigimod, on which he was lead investigator, employed a cyclic dosing schedule – 4 weeks’ treatment, then observation until patients needed retreatment — because patients demanded it.12 In clinical practice, some patients have gone more than 25 weeks before needing retreatment. One of his patients went beyond 40 weeks. “Others only get around 6-9 weeks. So patient choice again enters the decision-making process.”

Rituximab targets the CD20 protein on B cells nonspecifically, producing general immunosuppression. “That’s problematic in producing significant immunosuppression,” said Dr. Kaminski. Nevertheless, he said, rituximab is very effective for most patients with MuSK-specific MG, and its application to this indication has revealed differences between the MuSK subtype and AChR antibody–positive myasthenia. Specifically, MuSK antibody–positive patients have short-lived plasmablasts, which rituximab eliminates.13

Conversely, said Dr. Kaminski, patients with AChR antibody-positive myasthenia, especially long-term, likely have long-lived plasmablasts producing antibodies. This fact, and these patients’ lack of CD20, likely explain their poor response to rituximab.

A phase 3 trial (NCT04524273) of the CD19 blocker inebilizumab (Uplinza, Amgen) reached primary completion in May. Dr. Howard said that if topline results (unreleased at press time) prove positive, inebilizumab could replace rituximab in MG — provided payers do not reject inebilizumab because of cost.

 

 


Packed Early-Development Pipeline

Regarding early-stage projects, said Dr. Howard, the pipeline is packed with compounds that target various aspects of the immune system. “The real question with those is, what’s going to be the side effect profile? All of the trials are very early. We need bigger trials with much longer observation for safety, durability, and degree of efficacy.”

The next potential B cell–targeting game changer, he said, is chimeric antigen receptor (CAR) T cell–based therapy. In a phase 2b trial of Descartes-08 (Cartesian Therapeutics), 71% of treated patients experienced clinically meaningful improvement in MG Composite score at 3 months vs 25% for placebo.14

In early clinical trials, said Dr. Howard, patients treated with Descartes-08 — which uses autologous mRNA to target B-cell maturation antigen — have shown “exceptional improvement” lasting 20 or more months. Because the drug is not ingrained permanently into the genome, Descartes-08 avoids potentially severe side effects of DNA-targeting CAR T candidates. Dr. Howard hopes a phase 3 trial will commence around January 2025.

The tolerance approach exemplified by CNP-106 (COUR Pharmaceuticals) and a myasthenia gravis tolerogen (Toleranzia) seeks to prevent the immune system from recognizing and reacting to the NMJ abnormalities that produce myasthenia gravis, potentially providing a cure. “We look forward to those trials as they come online in the next 1-2 years,” said Dr. Howard.
 

Unmet Needs

Historically, neurologists believed that all myasthenia gravis symptoms stemmed from muscle fatigue — the more active the muscle, the weaker it gets. However, said Dr. Kaminski, some patients might lack measurable weakness but still complain of fatigue.

Elevated levels of cytokines such as interleukin (IL)–6 or IL-17 also can produce fatigue, he noted. “With the drugs we’re using, certainly the new ones, we’re not specifically targeting this fatigue phenomenon, which has been studied in a very limited fashion.”

In the RAISE-XT zilucoplan trial, participants experienced significant improvement in fatigue scores for up to 60 weeks.15 Although zilucoplan does not address fatigue directly, said Dr. Howard, improving myasthenia gravis overall helps reduce fatigue.

The Myasthenia Gravis Symptoms Patient Reported Outcome (MG Symptoms PRO), which Dr. Kaminski helped develop, includes questions designed to distinguish muscular fatigue from overall physical fatigue.16 “I’m very interested in some of the information that’s coming out on long COVID and its effect on muscle,” Dr. Kaminski added. “We might be able to learn from there that there’s still some pathology going on beyond the neuromuscular junction.”

What the field desperately needs, said Dr. Howard, are biomarkers to identify which patients will and will not respond to certain therapeutics. “We’re not there yet.” Such biomarkers are at least 3-7 years from becoming clinical reality.

Promising antibody-independent serum markers include circulating microRNAs. For example, miRNA-150-5p and miRNA-21-5p are elevated in generalized AChR-positive myasthenia gravis and early-onset myasthenia gravis (occurring before age 50) and decline after immunosuppression and thymectomy.17

Among diagnostic modalities for patients with seronegative myasthenia gravis, said Dr. Kaminski, single-fiber EMG is the most sensitive, at approximately 95%. “It’s not perfect.” Moreover, he said, performing this test accurately requires a highly experienced expert, which many treatment centers lack.

Presently, added Dr. Kaminski, orbital MRI is neither specific nor sensitive enough to be clinically useful. “One needs to be careful with these specialized tests that are published from the best laboratory in the world that does the test, and does it repetitively.” As the search for effective myasthenia gravis biomarkers continues, avoiding false-positive results is as important as avoiding false negatives.

 

References

1. Bubuioc AM et al. J Med Life. 2021 Jan-Mar;14(1):7-16. doi: 10.25122/jml-2020-0145.

2. Deenen JC et al. J Neuromuscul Dis. 2015;2(1):73-85. doi: 10.3233/JND-140045.

3. Kaminski HJ et al. J Clin Invest. 2024 Jun 17;134(12):e179742. doi: 10.1172/JCI179742.

4. Howard JF Jr et al. Lancet Neurol. 2017 Dec;16(12):976-986. doi: 10.1016/S1474-4422(17)30369-1.

5. Huda R. Front Immunol. 2020 Feb 21:11:240. doi: 10.3389/fimmu.2020.00240.

6. Howard JF Jr et al. Lancet Neurol. 2023 May;22(5):395-406. doi: 10.1016/S1474-4422(23)00080-7.

7. Vu T et al. NEJM Evid. 2022 May;1(5):EVIDoa2100066. doi: 10.1056/EVIDoa2100066.

8. Tice JA et al. October 20, 2021. https://icer.org/assessment/myasthenia-gravis/.

9. Yin J et al. Eur J Neurol. 2024 Aug;31(8):e16322. doi: 10.1111/ene.16322.

10. Antozzi C et al. EAN 2024, Abstract EPR-116. https://www.neurology.org/doi/10.1212/WNL.0000000000203660.

11. Yan C et al. JAMA Neurol. 2024 Mar 4;81(4):336-345. doi: 10.1001/jamaneurol.2024.0044.

12. Howard JF Jr et al. Lancet Neurol. 2021 Jul;20(7):526-536. doi: 10.1016/S1474-4422(21)00159-9.

13. Stathopoulos P et al. JCI Insight. 2017 Sep 7;2(17):e94263. doi: 10.1172/jci.insight.94263.

14. Cartesian Therapeutics. Cartesian Therapeutics announces positive topline results from phase 2b trial of Descartes-08 in patients with myasthenia gravis. 2024 Jul 2. https://ir.cartesiantherapeutics.com/news-releases/news-release-details/cartesian-therapeutics-announces-positive-topline-results-phase.

15. Howard JF Jr et al. Ther Adv Neurol Disord. 2024 Apr 17:17:17562864241243186. doi: 10.1177/17562864241243186.

16. Cleanthous S et al. Orphanet J Rare Dis. 2021 Oct 30;16(1):457. doi: 10.1186/s13023-021-02064-0.

17. Sabre L et al. Front Immunol. 2020 Mar 4:11:213. doi: 10.3389/fimmu.2020.00213.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article