A Patient Story: My Journey to a Better Life

By Mayra D.

My Primary Myelofibrosis journey started on a festive Cinco de Mayo in 2015 while I was at work. Without any warning, I started to feel sick to the point that I almost collapsed at my desk. I still remember this incident like it was yesterday. I was sweating and had an awful pain in my lower back that kept me out of my chair. I had very severe discomfort in my upper abdomen that was not an ordinary stomach pain. At that moment I knew I was struggling health-wise. I was feeling so weak and so out of balance, things I had never felt or experienced before in my life. After this scary episode, I was rushed to the ER. When I got to the hospital, I was not able to walk, and breathing was very difficult. Even holding my cell phone was a challenge. I was admitted right away due to the fact that my hemoglobin was very low among other things. After a bunch of blood transfusions, IV treatments, and everything in between, the doctors were able to stabilize my condition. After a week I was released from the hospital and started what I called ‘My Health Crisis Path’.


I started to see an Oncologist/Hematologist on a regular basis. One year later, I was rushed to the ER once again. At first the doctors thought I was pre-menopausal and that was the cause of the anemia.  No, it wasn’t!  The severe night sweats, the awful skin itching, especially after taking a shower, the painful feet, and leg cramps at night while in bed, and the severe anemia, were all part of a few of the new symptoms I was experiencing. Overall, my health was not improving, and I was feeling weaker every day. After a year and a half of trying new treatments with no diagnosis, the Oncologist/Hematologist team decided that the next step for me was a bone marrow biopsy. Everything became crystal clear with my ‘Jak 2 Mutation’ and the finding of Primary Myelofibrosis (MF).  

 

This health crisis of mine impacted me and my entire family.  My husband got so overwhelmed he even lost his job. I was forced to resign from my job and my husband losing his caused a great struggle financially. It was okay, and we managed together as a family, but having him by my side every step of the way was more important and something money can’t buy.

 

On the other hand, my daughter, who was only 14, felt like she was losing her mom. She reached rock bottom not only emotionally but academically.  Today I can say joyfully my daughter is now 21 years old and is a successful college student attending a State University. Through all of this, we saw a psychiatrist and had a few counseling sessions in order to help us individually and as a family cope with this unexpected health crisis. It has helped us with the healing and moving forward process.  Our daughter once said: “We are a family no one left behind.”

 

I used to donate blood and today I am on the other side of the chair. My driver’s license even says that I am an organ donor. Slowly but surely, I started to accept the fact that I suffered from a rare chronic blood disorder. I had two options in front of me, either feel sorry for myself and do nothing about it or go out there and make a difference. I chose the second option. It was not an easy task. In the beginning, I felt alone and confused, and I didn’t know where to go for help, support, and answers. When I heard the word ‘cancer’ for the first time it was a feeling I can’t explain. But this didn’t stop me. 

 

Being a former graphic designer, I was used to putting on a creative hat, so I knew what to do next. I began by researching about MPN in general. In the process, I came up with what I called ‘My 4 Daily Elements’: Chemo-Tablet Treatment, Anti-Inflammatory Nutritional Meal Plan, Routine of Exercise, and Mindfulness (I want to point out that when I started this health journey there was not much guidance like there is today). A few years ago, I received a couple of certificates in Modeling and Acting from a local school, so I decided to take advantage and use these tools. This knowledge and experience are my platform for cancer awareness. Today I’m proud to say I’m a member of a Cancer Support Community at the cancer institute in Orlando, FL. I’m also a member of MPN groups on social media. I was interviewed a couple of years ago and an article of my journey was published in Prevention Magazine. What an honor it was being able to speak about this rare and chronic blood disorder. We may be a small group, but our voices can make a big difference.


I am also the voice in the Latino community. I spoke on a local radio station where I was able to bring awareness to Central Florida. In my message I let the listeners understand that talking about cancer doesn’t make you a victim or a weak person, it makes us stronger by informing others. I even asked one of the CSC-Mental Health Therapist to join me for another live radio chat so we could talk about integrative medicine and what it offers the cancer community.

 

Next month, I’m going to attend my first meeting as part of the Patient and Family Advisory Council at Orlando Health. In December I’m also going to be participating in the Sea World 3 Mile Reindeer Run for pediatric cancer and bone marrow transplant programs at AdventHealth for the second year. Sharing my story is very important to me. If I can shed light, hope, and support to others then my mission is accomplished. “ME with a Purpose.”

 

Incyte Announces FDA Approval Of Pemazyre® (Pemigatinib) As The First And Only Targeted Treatment For Myeloid/Lymphoid Neoplasms (MLNs) With FGFR1 Rearrangement

August 26, 2022 at 10:00am EDT

 

 This marks the second indication for Pemazyre, which received accelerated FDA approval in 2020 for adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with a fibroblast growth factor receptor 2 (FGFR2) fusion or other rearrangement

 Pemazyre is the only FGFR inhibitor with multiple indications

WILMINGTON, Del.–(BUSINESS WIRE)–Aug. 26, 2022– Incyte (Nasdaq:INCY) today announced that the U.S. Food and Drug Administration (FDA) has approved Pemazyre® (pemigatinib), a selective fibroblast growth factor receptor (FGFR) inhibitor, for the treatment of adults with relapsed or refractory myeloid/lymphoid neoplasms (MLNs) with FGFR1 rearrangement. MLNs with FGFR1 rearrangement are extremely rare and aggressive blood cancers that may impact less than 1 in 100,000 people in the United States1.

“The approval of Pemazyre represents an important treatment advancement for people living with MLNs with FGFR1 rearrangement who currently have limited treatment options,” said Hervé Hoppenot, Chief Executive Officer, Incyte. “These are complex hematologic malignancies with a range of presentations, and this approval highlights Incyte’s continued leadership and commitment to advancing care for patients with rare blood cancers.”

A patient with an MLN with FGFR1 rearrangement may present with bone marrow involvement with a chronic myeloid malignancy (such as myeloproliferative neoplasm [MPN], myelodysplastic syndrome/MPN) or a blast phase malignancy (such as B- or T-cell acute lymphoblastic leukemia/lymphoma, acute myeloid leukemia or mixed phenotype acute leukemia). Bone marrow involvement may or may not be accompanied by extramedullary disease (EMD); some patients may present with EMD only. MLNs with FGFR1 rearrangement are caused by chromosomal translocations involving the FGFR1 gene, with various partner genes resulting in constitutive activation of the FGFR1 receptor tyrosine kinase, impacting cell differentiation, proliferation and survival2. Patients often relapse because existing first-line therapies sometimes fail to induce durable clinical and cytogenetic responses.

The FDA approval was based on data from the Phase 2 FIGHT-203 study, a multicenter open-label, single-arm trial that evaluated the safety and efficacy of Pemazyre in 28 patients with relapsed or refractory MLNs with FGFR1 rearrangement. Patients could have relapsed after allogeneic hematopoietic stem cell transplantation (allo-HSCT) or after a disease modifying therapy or were not a candidate for allo-HSCT or other disease modifying therapies.

  • Study participants included patients with documented MLNs with an 8p11 translocation on conventional cytogenetics and/or an FGFR1 rearrangement on break-apart FISH testing. (An FDA-approved test for detection of FGFR1 rearrangement in patients with relapsed or refractory MLNs is not available.)
  • In patients with chronic phase in the marrow with or without EMD (N = 18), the complete response (CR) rate was 78% (14/18; 95% CI 52, 94). The median time to response of CR was 104 days (range, 44 to 435 days). The median duration of CR was not reached (range, 1+ to 988+ days).
  • In patients with blast phase in the marrow with or without EMD (N = 4), two patients achieved a CR (duration: 1+ and 94 days).
  • In patients with EMD only (N = 3), one patient achieved a CR (duration: 64+ days).
  • For all patients (N = 28 including three patients without evidence of morphologic disease) the complete cytogenetic response rate was 79% (22/28; 95% CI: 59, 92).

The most common (≥ 20%) adverse reactions were hyperphosphatemia (74%), nail toxicity (62%), alopecia (59%), stomatitis (53%), diarrhea (50%), dry eye (50%), fatigue (44%), rash (35%), abdominal pain (35%), anemia (35%), constipation (32%), dry mouth (32%), epistaxis (29%), retinal pigment epithelial detachment (26%), extremity pain (26%), decreased appetite (24%), dry skin (24%), dyspepsia (24%), back pain (24%), nausea (21%), blurred vision (21%), peripheral edema (21%) and dizziness (21%).

“In patients with relapsed or refractory MLNs with FGFR1 rearrangement treated with Pemazyre in FIGHT-203, the high rate of complete response and complete cytogenetic response in patients with chronic phase disease and the high rate of complete cytogenetic response in patients with blast phase disease is clinically meaningful, especially in light of the lack of these specific responses with existing first-line treatments,” said Dr. Srdan Verstovsek, M.D., Ph.D., Professor, Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, and principal investigator for the FIGHT-203 study.

The supplemental New Drug Application (sNDA) for Pemazyre for the treatment of adults with relapsed or refractory MLNs with FGFR1 rearrangement was reviewed by the FDA under Priority Review. The FDA grants Priority Review to medicines that may offer a major advance in treatment where none currently exists. The designation shortens the review period to six months compared to 10 months for Standard Review.

Incyte established its leadership in rare blood cancers more than 10 years ago with the development of the first JAK inhibitor approved by the FDA for the treatment of certain patients with myelofibrosis and polycythemia vera. Incyte continues to research additional pathways to address rare blood cancers through its LIMBER (Leadership In MPNs Beyond Ruxolitinib) clinical development program, designed to evaluate multiple therapies and investigational strategies to improve and expand treatments for patients living with MPNs and other related hematologic malignancies and conditions.

Incyte is committed to supporting patients and removing barriers to access medicines. Eligible patients in the U.S. who are prescribed Pemazyre have access to IncyteCARES (Connecting to Access, Reimbursement, Education and Support), a comprehensive program offering personalized patient support, including financial assistance and ongoing education and additional resources. More information about IncyteCARES is available by visiting www.incytecares.com or calling 1-855-452-5234.

About FIGHT-203
FIGHT-203 is a Phase 2, multicenter trial that enrolled patients 18 years and older with myeloid/lymphoid neoplasms (MLNs) with a fibroblast growth factor receptor 1 (FGFR1) rearrangement. Sponsored by Incyte, the study evaluated the safety and efficacy of pemigatinib for the treatment of adults with MLNs with FGFR1 rearrangement. Patients received pemigatinib 13.5 mg once daily in 21-day cycles, either on a continuous schedule (the approved recommended starting dosage for use in patients with MLNs with FGFR1 rearrangement) or as an intermittent schedule (14 days on, 7 days off, an unapproved dosage regimen in MLN with FGFR1 rearrangement). Pemigatinib was administered until disease progression or unacceptable toxicity or until patients were able to receive allo-HSCT. For more information about the study, please visit https://clinicaltrials.gov/ct2/show/NCT03011372.

About Pemazyre® (pemigatinib)
Pemazyre, a fibroblast growth factor receptor (FGFR) inhibitor, is the first targeted treatment approved for use in the United States for treatment of adults with relapsed or refractory myeloid/lymphoid neoplasms (MLNs) with FGFR1 rearrangement.

Pemazyre is also indicated for the treatment of adults with relapsed or refractory previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with a FGFR2 fusion or other rearrangement as detected by an FDA-approved test. This indication is approved under accelerated approval based on overall response rate and duration of response (DOR). Continued approval may be contingent on verification and description of clinical benefit in a confirmatory trial(s).

IMPORTANT SAFETY INFORMATION

Ocular Toxicity
Retinal Pigment Epithelial Detachment (RPED): PEMAZYRE can cause RPED, which may cause symptoms such as blurred vision, visual floaters, or photopsia. Clinical trials of PEMAZYRE did not conduct routine monitoring including optical coherence tomography (OCT) to detect asymptomatic RPED; therefore, the incidence of asymptomatic RPED with PEMAZYRE is unknown.

Among 635 patients who received a starting dose of PEMAZYRE 13.5 mg across clinical trials, RPED occurred in 11% of patients, including Grade 3-4 RPED in 1.3%. The median time to first onset of RPED was 56 days. RPED led to dose interruption of PEMAZYRE in 3.1% of patients, and dose reduction and permanent discontinuation in 1.3% and in 0.2% of patients, respectively. RPED resolved or improved to Grade 1 levels in 76% of patients who required dosage modification of PEMAZYRE for RPED.

Perform a comprehensive ophthalmological examination including OCT prior to initiation of PEMAZYRE and every 2 months for the first 6 months and every 3 months thereafter during treatment. For onset of visual symptoms, refer patients for ophthalmologic evaluation urgently, with follow-up every 3 weeks until resolution or discontinuation of PEMAZYRE. Modify the dose or permanently discontinue PEMAZYRE as recommended in the prescribing information for PEMAZYRE.

Dry Eye: Among 635 patients who received a starting dose of PEMAZYRE 13.5 mg across clinical trials, dry eye occurred in 31% of patients, including Grade 3-4 in 1.6% of patients. Treat patients with ocular demulcents as needed.

Hyperphosphatemia and Soft Tissue Mineralization
PEMAZYRE can cause hyperphosphatemia leading to soft tissue mineralization, cutaneous calcification, calcinosis, and non-uremic calciphylaxis. Increases in phosphate levels are a pharmacodynamic effect of PEMAZYRE. Among 635 patients who received a starting dose of PEMAZYRE 13.5 mg across clinical trials, hyperphosphatemia was reported in 93% of patients based on laboratory values above the upper limit of normal. The median time to onset of hyperphosphatemia was 8 days (range 1-169). Phosphate lowering therapy was required in 33% of patients receiving PEMAZYRE.

Monitor for hyperphosphatemia and initiate a low phosphate diet when serum phosphate level is >5.5 mg/dL. For serum phosphate levels >7 mg/dL, initiate phosphate lowering therapy and withhold, reduce the dose, or permanently discontinue PEMAZYRE based on duration and severity of hyperphosphatemia as recommended in the prescribing information.

Embryo-Fetal Toxicity
Based on findings in an animal study and its mechanism of action, PEMAZYRE can cause fetal harm when administered to a pregnant woman. Oral administration of pemigatinib to pregnant rats during the period of organogenesis caused fetal malformations, fetal growth retardation, and embryo-fetal death at maternal exposures lower than the human exposure based on area under the curve (AUC) at the clinical dose of 13.5 mg.

Advise pregnant women of the potential risk to the fetus. Advise female patients of reproductive potential to use effective contraception during treatment with PEMAZYRE and for 1 week after the last dose. Advise males with female partners of reproductive potential to use effective contraception during treatment with PEMAZYRE and for 1 week after the last dose.

Adverse Reactions: Cholangiocarcinoma
Serious adverse reactions occurred in 45% of patients receiving PEMAZYRE (n=146). Serious adverse reactions in ≥2% of patients who received PEMAZYRE included abdominal pain, pyrexia, cholangitis, pleural effusion, acute kidney injury, cholangitis infective, failure to thrive, hypercalcemia, hyponatremia, small intestinal obstruction, and urinary tract infection. Fatal adverse reactions occurred in 4.1% of patients, including failure to thrive, bile duct obstruction, cholangitis, sepsis, and pleural effusion.

Permanent discontinuation due to an adverse reaction occurred in 9% of patients who received PEMAZYRE. Adverse reactions requiring permanent discontinuation in ≥1% of patients included intestinal obstruction and acute kidney injury.

Dosage interruptions due to an adverse reaction occurred in 43% of patients who received PEMAZYRE. Adverse reactions requiring dosage interruption in ≥1% of patients included stomatitis, palmar-plantar erythrodysesthesia syndrome, arthralgia, fatigue, abdominal pain, AST increased, asthenia, pyrexia, ALT increased, cholangitis, small intestinal obstruction, alkaline phosphatase increased, diarrhea, hyperbilirubinemia, electrocardiogram QT prolonged, decreased appetite, dehydration, hypercalcemia, hyperphosphatemia, hypophosphatemia, back pain, pain in extremity, syncope, acute kidney injury, onychomadesis, and hypotension.

Dose reductions due to an adverse reaction occurred in 14% of patients who received PEMAZYRE. Adverse reactions requiring dosage reductions in ≥1% of patients who received PEMAZYRE included stomatitis, arthralgia, palmar-plantar erythrodysesthesia syndrome, asthenia, and onychomadesis.

Clinically relevant adverse reactions occurring in ≤10% of patients included fractures (2.1%). In all patients treated with pemigatinib, 0.5% experienced pathologic fractures (which included patients with and without cholangiocarcinoma [N = 635]). Soft tissue mineralization, including cutaneous calcification, calcinosis, and non-uremic calciphylaxis associated with hyperphosphatemia were observed with PEMAZYRE treatment.

Within the first 21-day cycle of PEMAZYRE dosing, serum creatinine increased (mean increase of 0.2 mg/dL) and reached steady state by Day 8, and then decreased during the 7 days off therapy. Consider alternative markers of renal function if persistent elevations in serum creatinine are observed.

In cholangiocarcinoma (n=146) the most common adverse reactions (incidence ≥20%) were hyperphosphatemia (60%), alopecia (49%), diarrhea (47%), nail toxicity (43%), fatigue (42%), dysgeusia (40%), nausea (40%), constipation (35%), stomatitis (35%), dry eye (35%), dry mouth (34%), decreased appetite (33%), vomiting (27%), arthralgia (25%), abdominal pain (23%), hypophosphatemia (23%), back pain (20%), and dry skin (20%).

Adverse Reactions: Myeloid/Lymphoid Neoplasms with FGFR1 Rearrangement

Serious adverse reactions occurred in 53% of patients receiving PEMAZYRE at all dosages (n=34). Serious adverse reactions in > 5% of patients included acute kidney injury. Fatal adverse reactions occurred in 9% of patients who received PEMAZYRE, including acute kidney injury, multiple organ dysfunction syndrome, and malignant neoplasm progression, occurring in one patient each.

Permanent discontinuation due to an adverse reaction occurred in 12% of patients who received PEMAZYRE at all dosages. Adverse reactions requiring permanent discontinuation included cardiac failure, multiple organ dysfunction syndrome, blood alkaline phosphatase increase, and calciphylaxis. In patients who started treatment on the recommended dosage (n = 20), adverse reactions requiring dosage interruption of PEMAZYRE occurred in 80% of patients. Adverse reactions which required dosage interruption in > 2 patients treated at the recommended dosage included nail toxicities (20%) and hyperphosphatemia (15%).

Dose reductions of PEMAZYRE due to an adverse reaction occurred in 80% of patients who started treatment on the recommended dosage. Adverse reactions requiring dose reductions occurring in > 2 patients were nail toxicities (20%), hyperphosphatemia (20%), and alopecia (15%).

The most common (≥ 20%) adverse reactions were hyperphosphatemia (74%), nail toxicity (62%), alopecia (59%), stomatitis (53%), diarrhea (50%), dry eye (50%), fatigue (44%), rash (35%), abdominal pain (35%), anemia (35%), constipation (32%), dry mouth (32%), epistaxis (29%), retinal pigment epithelial detachment (26%), extremity pain (26%), decreased appetite (24%), dry skin (24%), dyspepsia (24%), back pain (24%), nausea (21%), blurred vision (21%), peripheral edema (21%), and dizziness (21%).

Drug Interactions

Avoid concomitant use of strong and moderate CYP3A inhibitors with PEMAZYRE. Reduce the dose of PEMAZYRE if concomitant use with a strong or moderate CYP3A inhibitor cannot be avoided. Avoid concomitant use of strong and moderate CYP3A inducers with PEMAZYRE.

Special Populations
Advise lactating women not to breastfeed during treatment with PEMAZYRE and for 1 week after the last dose.

Reduce the recommended dose of PEMAZYRE for patients with severe renal impairment as described in the prescribing information.

Reduce the recommended dose of PEMAZYRE for patients with severe hepatic impairment as described in the prescribing information.

Please see Full Prescribing Information for PEMAZYRE.

You are encouraged to report negative side effects of prescription drugs to the FDA. Visit www.fda.gov/medwatch, or call 1-800-FDA-1088.

You may also report side effects to Incyte Medical Information at 1-855-463-3463.

 

Read more

Paving the way to improve therapy for Myeloproliferative Neoplasms

Megan Bywater & Steven W. Lane

Nature Communications 13, Article Number: 5052 (2022)

 

Long-acting IFNα induces durable molecular responses in myeloproliferative neoplasms. Emerging studies, including Saleiro et al. recently published in Nature Communications, have identified promising candidates that may synergise with IFNα by targeting stem cell function or feedback loops that mediate treatment resistance.

Clinical management in Myeloproliferative Neoplasms

The MyeloProliferative Neoplasms (MPN) are characterised by the excess production of phenotypically normal mature myeloid cells or cell products, specifically red blood cells in polycythemia vera (PV), megakaryocytes and platelets in essential thrombocythemia (ET) and additional cytokine driven fibro-cellular infiltration of the bone marrow in primary myelofibrosis (PMF)1. This expansion of mature myeloid cell populations is driven by the constitutive activation of the JAK-STAT signalling pathway in committed myeloid progenitors as a consequence of mutations in either JAK2MPL or CALR1. However, although this disease is phenotypically driven from expanded committed myeloid progenitor cell compartments, these populations remain untransformed as they lack the capacity for long-term self-renewal. As a consequence, MPN-driving mutations must be maintained within the haematopoietic stem cell (HSC) compartment2.

MPNs are long-term chronic conditions and patient management is focussed on ameliorating the symptoms related to clinical pathologies1. Current approaches are limited by toxicity of long-term treatments that have little disease modifying activity and do not prevent transformation to more aggressive diseases such as leukaemia. Venesection leads to iron restricted erythropoiesis and is intended to reduce the expanded mature red blood cell population. To a similar end, hydroxycarbamide (also known as hydroxyurea) is frequently used for cytoreduction to control erythrocytosis and thrombocytosis with a concomitant effect on reducing thrombotic tendency. More recently, small molecule inhibitors of JAKs have been developed to target the signalling pathway hyperactivated in this disease. Unfortunately, current studies indicate that the MPN stem cell pool is not reliant on the constitutive activation of JAK2 for survival3. Consequently, JAK1/2 inhibitors like Ruxolitinib have proven effective at reducing the excess production of mature myeloid cells, inflammatory cytokine levels and the associated clinical symptoms in MPN, but have had limited efficacy in reducing the size of the MPN stem cell pool3. Therefore, long-term disease management via targeted JAK2 inhibition will most likely require chronic administration of these compounds, the practicality of which can be limited by significant side effects.

Disease progression to secondary myelofibrosis (sMF) or acute myeloid leukaemia (sAML) occurs in ~8–20% and 8–26% of patients with ET and PV respectively over a 20-year period1 and is related to an expanded mutational spectrum4. Importantly, clinical outcomes subsequent to disease progression are poor on account of limited effective treatment options. In the case of sMF and MPN-driver-positive sAML, it is assumed that transformation is driven by the selective pressure provided by the MPN-driving oncogene. Considering this, it would seem the most rational approach for the clinical management of MPN is the development of treatment options that selectively target the MPN stem cell pool to decrease both the burden of the chronic management of this disease and prevent the deleterious outcomes related to disease progression.

Long-acting IFNα induces durable responses in MPN

Recent years have seen a renaissance in the use of interferon alpha (IFNα) for the treatment of MPN, specifically PV and ET. Initially, the use of IFNα was rationalised on the basis of its known myelosuppressive effects with several groups also postulating an immunostimulatory role of these agents. The clinical uptake of IFNα therapy was initially hampered by low compliance related to a poor pharmacokinetic profile of unmodified recombinant forms. More recently, pegylated versions of IFNα have been found to be more persistent in vivo, extending the duration of response and allowing a longer interval between doses. Several studies have now compared long-acting pegylated IFNα with hydroxycarbamide in inducing durable long-term haematological responses5,6,7, including the normalisation of red blood cell counts and the prevention of thromboembolic events, with the advantage of also being non-leukemogenic. Importantly, IFNα therapy has also proven effective at targeting the MPN stem cell pool with durable molecular remissions also being observed across multiple studies5,6,7.

Mechanistically, IFNα drives cell cycle entry in HSCs with this mitogenic effect being more potent in Jak2-mutant HSCs3,8 supporting a prevailing hypothesis that molecular remissions observed in PV patients receiving IFNα are due to the preferential functional decline of the MPN stem cell pool. Despite its appreciable clinical success, it is clear that the selectivity of IFNα for mutant MPN stem cells over normal stem cells is mild. Consequently, there is an active field of interest looking to understand this selectivity and exploit it through combination therapies. Recently published in Nature Communications, Saleiro et al.9 further elucidates the ability of IFNα to activate a PKCd-ULK1-p38 MAPK signalling cascade that acts in parallel to STAT1 to drive transcription of interferon response genes (IRGs) and that genetic disruption of this pathway can attenuate the ability of IFNα to reduce self-renewal in malignant erythroid precursors. In demonstrating that ULK1 preferentially associates with the activated forms of ROCK1/2 and that IFNα also drives ROCK1/2 activation, they postulate whether modulating ROCK1/2 activity directly may also affect cellular responses to IFNα. In support of this, they demonstrate that both genetic and pharmacological inhibition of ROCK1/2 can enhance the ability of IFNα to reduce cell viability in JAK2-mutant cell lines and self-renewal in PV patient erythroid precursors9. It will be important to determine whether this combination will have enhanced selectivity in the targeting of MPN stem cells over normal stem cells.

Another intriguing combinatorial approach to enhance MPN stem cell selectivity exploits the higher basal levels of PML-nuclear bodies (NB) present in JAK2V617F HSCs10. Arsenic trioxide (ATO) is the standard of care in acute promyelocytic leukaemia and acts in part through degradation of the driving oncogene PML/RAR alpha. ATO can also drive PML-NB formation, which has been shown to be tumour suppressive. Notably, PML is also an IRG, with the combination of IFNα and ATO proving highly effective in preferentially increasing PML-NB formation in Jak2-mutant stem cells and reducing their capacity to transplant disease10. Perhaps counterintuitive is the combination of IFNα with the JAK1/2 inhibitor Ruxolitinib, which clearly has clinical activity, although trials have been limited by toxicity11. Although both agents have potent activity against JAK2V617F MPN, JAK1 kinase activity is required for IFNα-mediated phosphorylation and activation of STAT1. Interestingly, despite being able to robustly inhibit STAT1 phosphorylation in LT-HSCs in response to IFNα in vitro, Ruxolitinib effects only minimal attenuation of STAT1 phosphorylation and cell cycle entry of LT-HSCs in response to IFNα in vivo3. These data suggest this combination may be able to exploit the MPN stem cell-selective effects of IFNα in addition to the anti-proliferative and anti-inflammatory effects of Ruxolitinib on MPN myeloid precursors and committed cells.

Evolving resistance to IFNα mediated by alternate signalling pathways

Saleiro et al.9 also postulate the utility of identifying biomarkers of IFNα treatment response in the clinical management of MPN. They show that the hyperactivation of the PKCd-ULK1-p38 MAPK pathway may enhance the therapeutic response to IFNα treatment in MPN, in that increased expression of ULK1 and p38 MAPK mRNA correlates with haematological responses in a combined cohort of IFNα-treated PV and ET patients. Consistent with this, a number of genetic determinants of IFNα treatment response are emerging. Notably, despite the convergent mechanistic pathways of MPN-driver mutations, it appears that the ability of IFNα to deplete the MPN stem cell clone is largely restricted to patients with JAK2V617F mutations, and that CALR mutant MPN is less likely to achieve molecular remissions in response to pegylated IFNα therapy, despite achieving similar outcomes in terms of haematological responses12,13. Early studies also indicate that DNMT3A mutations are enriched after IFNα treatment, suggesting a possible association of this mutation with IFNα resistance in patients14. As such, we must consider how molecular responses to MPN therapies may be modified by the growing list of concomitant mutations in this disease4,15. To achieve this it is imperative that recent large cohort studies comparing the efficacy of IFNα therapy to standard of care should be combined with Next Generation Sequencing analysis of pathogenic loci, to determine what concomitant mutations are associated with IFNα treatment outcomes. This is important as, in the long-term, concomitant mutations in genes, including TP53EZH2 and ASXL1, confer a higher risk of disease transformation to phenotypes with poor clinical outcomes, such as MF and AML1. Consequently, these studies are vital for determining the potential of IFN therapy to delay or prevent disease progression.

Conclusions

In summary, the clinical management of patients with MPN has been dominated by three main approaches: chemotherapy for cytoreduction, targeted JAK2 inhibition or long-acting IFNα analogues. Although all are highly effective, to date none have shown the sustained ability to modify the natural history of disease and prevent transformation to sAML or sMF. Long-acting pegylated IFNα is the only therapy to show reliable and deep molecular responses, but requires long-term treatment and is often limited by toxicity. Studies, like Saleiro et al., have identified promising candidates that may synergise with IFNα by targeting stem cell function or feedback loops that mediate treatment resistance. The next phase of clinical studies should address rational combinations and the contribution of concomitant mutations to treatment response, with ambitious clinical endpoints, including molecular remission, treatment free remission and prevention of disease transformation.

Read more

Pelabresib and Ruxolitinib Combination Proves Durable in Patients With Myelofibrosis

Targeted Therapies in OncologyAugust 2022, Volume 11, Issue 11

Results from the MANIFEST study of pelabresib and ruxolitinib in myelfibrosis are positive. The phase MANIFEST-2 study continue to explored the combination in JAK inhibitor–naïve patients with myelofibrosis.

The combined use of pelabresib (CPI-0610) and ruxolitinib (Jakafi) demonstrated durable responses beyond week 24 in patients with myelofibrosis who experienced a suboptimal response to ruxolitinib and in those who were JAK inhibitor naïve, according to preliminary data from arms 2 and 3 of the phase 1/2 MANIFEST trial (NCT02158858).1

Findings, which were presented during the 2022 European Hematology Association Congress, showed that 68% (95% CI, 57%-78%) of patients with JAK inhibitor–naïve myelofibrosis (n = 84; arm 3) experienced a reduction in spleen volume of at least 35% (SVR35) at week 24 with the combination. Eighty percent of patients achieved this at any point in the study, and 69% of those with SVR35 maintained response at the time of data cutoff. Moreover, 56% (95% CI, 45%-67%) of patients in this arm experienced a 50% or greater reduction in total symptom score (TSS) from baseline at week 24, resulting in a median TSS change of –59% at this time point.

In patients with myelofibrosis who had previously experienced a suboptimal response to ruxolitinib (n = 81; arm 2), 20% experienced SVR35 at week 24; this was observed in 17% of those who were transfusion dependent (TD) and 26% of those who were non–transfusion dependent (NTD). Additionally, 30% of patients achieved SVR35 at any time point. A 25% or higher SVR from baseline by week 24 was noted in 27% of patients. The median SVR was –18%. The addition of pelabresib to ruxolitinib positively affected symptoms in these patients, as well. The TSS50 at week 24 was 37%; this rate was 36% in TD patients and 39% in NTD patients, with a median symptom burden reduction of –47%.

“The combination of pelabresib and ruxolitinib was generally well tolerated, and preliminary results showed durable improvements in splenomegaly and symptom burden, with associated biomarker results suggesting potential disease-modifying activity,” said Claire Harrison, MD, FRCP, FRCPath, lead study author and professor of myeloproliferative neoplasms at Guy’s and St. Thomas’ NHS Foundation Trust in London, England, during a presentation of the data.

Setting the Stage

The JAK inhibitor ruxolitinib serves as the current standard of care for patients with myelofibrosis who are not candidates for hematopoietic stem cell transplant. However, JAK inhibitors are associated with limited efficacy and high discontinuation rates due to toxicities. As such, a large unmet need remains for the treatment of these patients with this disease.

The BET inhibitor pelabresib downregulates the expression of genes that contribute to the heterogenous features of the pathology of myelofibrosis, including aberrant erythroid and megakaryocytic differentiation, according to Harrison. Preclinical data have suggested that the combination of BET and JAK inhibitors could have synergistic activity in this disease.

In the ongoing, global, open-label MANIFEST study, investigators sought to further evaluate pelabresib in patients with myelofibrosis and essential thrombocythemia. The trial was composed of 4 arms.

Arm 1 included patients with myelofibrosis who were no longer on ruxolitinib and who would receive pelabresib monotherapy in the second-line setting. Those in arm 4 had highrisk, essential thrombocythemia and were resistant to or intolerant of hydroxyurea (Hydrea).

Patients in arm 2 were given pelabresib as an “add on” to ruxolitinib; these patients had achieved a suboptimal response with ruxolitinib or experienced disease progression. Those in this arm were further divided into 2 cohorts: patients who were TD (n = 59; cohort 2A) and those who were NTD (n = 27; cohort 2B). The primary end point in cohort 2A was conversion of TD to transfusion independence (TI), and key secondary end points included SVR35 and TSS50. For cohort 2B, the primary end point was SVR35, and a key secondary end point was TSS50.

Patients enrolled in arm 3 did not previously receive a JAK inhibitor and were considered to have intermediate-2 or high-risk disease by the Dynamic International Prognostic Scoring System (DIPSS). These patients received the combination of pelabresib and ruxolitinib in the first-line setting. Here, the primary end point was SVR35 and the secondary end point was TSS50.

“Both these [groups of] patients have over 80 patients in them, and I just want to highlight that the vast majority were intermediate-2 or high-risk by DIPSS,” Harrison explained. “They represent a high-risk cohort, with more than 50% of patients—two-thirds on arm 3 and nearly 80% on arm 2—being anemic already. Spleen volumes were large and concerning and over half of patients in both arms had high molecular risk mutations.”

Delving Into the Data

Additional data showed that within arm 2, 16% of 38 patients in cohort 2A converted from TD to TI. Moreover, 22% of 27 patients in cohort 2B were found to experience a hemoglobin response, defined as a post-baseline mean increase of at least 1.5 g/dL, which is required for any 12 weeks’ red blood cell transfusion-free period.

Notably, durable SVR was observed over time in both arms 3 and 2. “In the JAK inhibitor–naïve patients, [we saw] progressive reduction in spleen volume,” Harrison noted. “Spleen volume reduction over time [was also observed] in the second-line [patients].”

For arm 3, the median treatment duration of pelabresib plus ruxolitinib was not yet reached (95% CI, 19.2-not reached), and the median follow-up time was 21.8 months (95% CI, 21.2-22.5). In arm 2, the median treatment duration with pelabresib add-on to ruxolitinib was 14.0 months (95% CI, 8.4- 20.6), and the median follow-up time was 24.4 months (95% CI, 23.0-30.7).

Additional analyses revealed improvements in bone marrow fibrosis grade following 24 weeks of treatment by central pathology review. Specifically, 28% of patients in arm 3 (n = 57) and 26% of those in arm 2 (n = 47) experienced an improvement of at least 1 grade at this time point. Notably, these responses proved to be durable, with 56% and 50% of patients, respectively, maintaining their bone marrow fibrosis improvement at the next available assessment or beyond. Overall, at any time in the study, 40% and 39% of patients, respectively, achieved at least a 1-grade improvement in bone marrow fibrosis.

Utilizing digital pathology, investigators further evaluated features of the bone marrow biopsy. Again, improvements in bone marrow fibrosis were observed, with an increase in erythrocytes and a decrease in megakaryocyte clusters.

“Clustering of megakaryocytes is a key pathological feature in this disease. We saw that over 40% of patients in arm 2 and over 50% of patients in arm 3 experienced a 15% reduction in clustering of megakaryocytes,” Harrison said. “Megakaryocyte ‘de-clustering’ in the bone marrow was correlated with SVR35 response.”

A cytokine analysis was performed on paired plasma samples, and investigators noted decreased plasma levels of myelofibrosis-associated or inflammation-related cytokines in patients enrolled in arms 2 and 3.

A reduction in JAK2 variant allele frequency (VAF) and bone marrow fibrosis observed with pelabresib plus ruxolitinib was seen in patients within arm 3. “[With regard to] JAK2 V617F VAF reduction, a 20% reduction was attained by over one-third of patients, at only week 24 in the study,” Harrison said. “Regardless of the baseline VAF, there was a reduction in JAK allele burden. [Bone marrow fibrosis grade change] occurred in patients regardless of whether they had a spleen volume response or not, but there seems to be a nice correlation across all these different dimensions of this disease.”

Safety Statistics

Regarding safety, in the JAK inhibitor–naïve patients, the most common hematologic treatment-emergent adverse effects (TEAEs) experienced with the combination were anemia (all grade, 42%; grade 3, 33%; grade 4, 1%) and thrombocytopenia (all grade, 52%; grade 3, 8%; grade 4, 4%). Gastrointestinal (GI) events were noted to be mostly low grade; they included diarrhea (all grade, 35%; grade 3, 1%), constipation (all grade, 25%), nausea (all grade, 24%), and abdominal pain (23%).

A total of 5 grade 5 TEAEs were reported, and they included COVID-19 (n = 1), multiorgan failure due to sepsis secondary to infections (n = 2), and acute respiratory distress syndrome because of ruxolitinib withdrawal (n = 2). The investigators determined that none were related to pelabresib except in the case of the patient who experienced multiorgan failure due to sepsis secondary to pneumonia.

Among those in arm 2 who had previously experienced a suboptimal response to ruxolitinib, serious AEs reported in 3 or more patients included anemia (n = 6), respiratory tract infections (n = 4), and urinary tract infections (n = 3). “Similar data were observed in this tougher cohort of patients in the second-line setting with thrombocytopenia and anemia representing grade 3 and 4 events,” Harrison said. Twenty patients experienced TEAEs that resulted in the discontinuation of pelabresib. “Grade 3 GI events are not common causes for discontinuation and are easily managed,” Harrison added. A total of 7 grade 5 TEAEs occurred in patients within this arm, and included acute kidney injury, traumatic subdural hematoma, brain stem hemorrhage, disease progression, transformation to acute myeloid leukemia, congestive heart failure, and suspected lung cancer. All TEAEs, except for acute kidney injury, were determined to not be associated with pelabresib.

The double-blind, phase 3 MANIFEST-2 trial (NCT04603495), which is evaluating pelabresib plus ruxolitinib compared with placebo and ruxolitinib in JAK inhibitor–naïve patients with myelofibrosis, has been initiated and is open for enrollment.

 

REFERENCE:

1. Mascarenhas J, Kremyanskaya M, Patriarca A, et al. BET inhibitor pelabresib (CPI-0610) combined with ruxolitinib in patients with myelofibrosis––JAK inhibitor-naïve or with suboptimal response to ruxolitinib––preliminary data from the MANIFEST study. Presented at: European Hematology Association 2022 Congress; June 9-12, 2022; Vienna, Austria. Abstract S198.

Read more

How to Treat Algodystrophy and Rheumatic Comorbidity in Myelofibrosis: Three Case Reports

Published: August 16, 2022 (see history)

DOI: 10.7759/cureus.28058

Cite this article as: Magazzino O, Urbano T, Magnasco S (August 16, 2022) How to Treat Algodystrophy and Rheumatic Comorbidity in Myelofibrosis: Three Case Reports. Cureus 14(8): e28058. doi:10.7759/cureus.28058

Abstract

Algodystrophy or complex regional pain syndrome is a chronic pain condition characterized by hyperalgesia and allodynia. Patients with algodystrophy present an amplified and persistent activation of the innate immune system, with subsequent proliferation of keratinocytes and release of proinflammatory cytokines including interleukin (IL)-6, IL-1β, and tumor necrosis factor-α (TNF-α). Chronic inflammation and increased levels of cytokines are observed also in Ph-negative myeloproliferative neoplasms, including polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Chronic myeloid neoplasms are characterized by overproduction of one or more mature non-lymphoid cell lineages, with erythrocytosis, thrombocytosis, and/or myeloproliferation.

Three case reports described our experience in the treatment of algodystrophy and rheumatic conditions in patients with myelofibrosis; a literature search was also performed.

The first patient was a 58-year-old woman who suffered from chronic myeloproliferative neoplasm in myelofibrotic evolution, under treatment with ruxolitinib and pre-treated with hydroxyurea; she reported inflammatory pain, and swelling of the tibiotarsal joints bilaterally. She was treated with neridronate 2 mg/kg for four days and methotrexate 15 mg per os per week, achieving a clinical benefit. The second patient was a 63-year-old woman diagnosed with polycythemia vera evolving to myelofibrosis. She experienced pain and swelling of the left tibiotarsal joint and difficulty walking. A therapy with low-dose steroid per os and intramuscular clodronate was administered for four months, followed by methotrexate at 15 mg per week. After two months, tenosynovitis significantly improved, as supported by the evidence of improved bone edema of the left tibiotarsal joint revealed in the magnetic resonance imaging, and pain symptoms were clinically ameliorated. The third patient was a 70-year-old male patient affected by essential thrombocythemia with myelofibrotic evolution and a paraneoplastic polymyalgia rheumatica treated with steroids and currently in remission. The patient received ruxolitinib for about two years; after the first year of treatment, he experienced pain and swelling of the right tibiotarsal joint with difficulty in walking, with a consequent diagnosis of edema and tenosynovitis, as per algodystrophy. After consulting a rheumatologist, the patient received therapy with neridronate intramuscularly with clinical benefit.

As overlapping interactions and clinical manifestations between hematologic neoplasms and rheumatologic diseases exist, new clinical manifestations, such as algodystrophy, may emerge during myelofibrosis and need to be monitored in the long term by a multidisciplinary team.

Introduction

Algodystrophy is a chronic pain condition characterized by hyperalgesia and allodynia that can develop after extremity trauma, infection, or surgery [1]. The main features of algodystrophy are abnormal tissue response to injury, sensitization of the peripheral and central nervous systems, inflammatory changes, and autonomic dysregulation [2].

Focusing on the underlying inflammatory process, the clinical course of algodystrophy consists of an acute or warm phase, in which pro-inflammatory modulators are released, and a chronic or cold phase, where keratinocytes, fibroblasts, and osteocytes are activated [2].

During the acute phase, the release of pro-inflammatory cytokines, including interleukin-6 (IL-6), IL-1β, and tumor necrosis factor-α (TNF-α) [3], triggers an immune cascade that results in histamine-induced vasodilation, causing the redness, swelling, pain, and warmth. These cytokines also activate the connective tissue, causing contractures [4], and alter bone metabolism by acting on osteoblasts and osteoclasts [5]. Then, during the chronic phase, rapid bone turnover, bone loss, and osteoporotic changes occur [5].

Some evidence in animal models and preclinical studies indicate that even autoimmunity plays a role in algodystrophy [6]. In mice treated with anti-CD20 and in mu-MT mice (lacking mature B cells), after fracture/cast immobilization, algodystrophy-like symptoms were less severe compared to wild-type mice that had undergone the same procedure; IgM deposition and complement activation were also observed in the skin and sciatic nerves of wild-type fracture/cast mice [7]. Furthermore, experiments using immunohistochemical techniques and fluorescence-assisted cell sorting (FACS) analysis identified sympathetic nervous system neurons as targets for autoantibodies in some patients with algodystrophy, with little evidence of such autoimmunity from patients with other types of peripheral neuropathy [8].

Algodystrophy shows a variable progression over time and early initiation of the therapy is mainly aimed at restoring limb functionality, decreasing pain, and improving the quality of life. To reach these goals, a multidisciplinary approach involving patient education, physical and occupational therapy, along with pharmacological and surgical interventions, is helpful.

Non-steroidal anti-inflammatory drugs (NSAID) and corticosteroids have been traditionally used to manage pain and inflammation of algodystrophy; furthermore, based on the positive results that emerged from small randomized clinical trials, bisphosphonates are also introduced in the treatment of algodystrophy [2]. Bisphosphonates can modulate inflammatory mediators, proliferation, and migration of bone marrow cells but their mechanism of action has not been accurately detailed. Over the past three decades, several case reports described positive results in controlling pain, local inflammation, functional disability, and improving the quality of life of patients, especially in patients with early disease [9]. A randomized trial compared the efficacy of neridronate versus placebo in patients with algodystrophy and showed a significant improvement in the indices of pain and quality of life [10]. A meta-analysis of four randomized clinical trials including a total of 181 patients showed a significant reduction of pain in patients with algodystrophy with bisphosphonates compared to placebo, demonstrating the efficacy and safety of bisphosphonates in the treatment of the disease [11].

As in algodystrophy, inflammation is considered one of the factors that contribute to the development and progression of Ph-negative myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF).

Indeed, current evidence suggests that MPNs are chronic inflammatory conditions in addition to neoplastic disorders and that both processes contribute to the clinical manifestations and pathogenesis of the disease [12]. The relationship between inflammation and myeloproliferation is supported by the evidence that increased levels of circulating cytokines and chemokines and the accumulation of reactive oxygen species in chronic inflammatory states can lead to genetic instability, which may promote the development and progression of neoplasms [12].

In MPNs, hyperactivation of the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling due to the activating mutation V617F in the Janus kinase (JAK) 2 gene is frequently observed; in polycythemia vera and essential thrombocythemia, the JAK2 mutation can sustain a condition of chronic inflammation, explaining the associated constitutional symptoms, thrombosis, and premature atherosclerosis observed in patients with these disorders [12].

As the activating mutation V617F is a driver mutation in MPNs and is present in approximately 50% of patients with myelofibrosis, ruxolitinib, a potent oral inhibitor of JAK1/2, was tested in patients with myelofibrosis to examine the potential clinical benefit of JAK inhibition in this patients [13]. In a phase 1/2 trial, ruxolitinib showed clinical benefits associated with a marked diminution of levels of circulating inflammatory cytokines [13]. Therapeutic JAK2 inhibition with ruxolitinib reduced plasma levels of multiple cytokines in patients with myelofibrosis within the first month of treatment, without reverting them, however, to the low levels seen in healthy control plasmas [14]. Therefore, ruxolitinib can provide a partial, but incomplete, reduction of inflammatory pathophysiology in myelofibrosis. Other drugs currently used in patients with MPNs are hydroxyurea, anagrelide, and interferon [15].

Considering the high similarity in the inflammatory pathogenesis underlying both algodystrophy and MPNs, it is expected that clinical manifestations of rheumatological disorders are not uncommon during hematological malignancies.

In these case reports, we described our experience in the treatment of algodystrophy and rheumatic conditions in patients with myelofibrosis.

Case Presentation

Per the World Medical Association Declaration of Helsinki, all the data referring to the patients are published anonymously, without any details allowing re-identification of the patient. Informed consents were signed by the patient, as required by the law of the country.

Read more

US FDA accepts new drug application for GSK’s momelotinib for the treatment of myelofibrosis

August 17, 2022

  • Regulatory submission included data from the pivotal MOMENTUM phase III clinical trial that met all primary and key secondary efficacy endpoints

GSK plc (LSE/NYSE: GSK) today announced that the US Food and Drug Administration (FDA) accepted the New Drug Application (NDA) for momelotinib, a potential new medicine with a proposed differentiated mechanism of action that may address the significant unmet medical needs of myelofibrosis patients with anaemia. The US FDA has assigned a Prescription Drug User Fee Act action date of 16 June 2023.

The NDA is based on the results from key phase III trials, including the pivotal MOMENTUM trial, which met all primary and key secondary endpoints, including Total Symptom Score (TSS), Transfusion Independence (TI) rate and Splenic Response Rate (SRR). The primary analysis data from the MOMENTUM trial were recently presented at the 2022 American Society of Clinical Oncology Annual Meeting and the European Hematology Association 2022 Hybrid Congress.

Momelotinib is not currently approved in any market.

About the pivotal MOMENTUM phase III clinical trial

MOMENTUM is a global, randomised, double-blind phase III clinical trial of momelotinib versus danazol in patients with myelofibrosis who were symptomatic and anaemic and had been previously treated with an FDA-approved JAK inhibitor. The trial was designed to evaluate the safety and efficacy of momelotinib for treating and reducing key hallmarks of the disease: symptoms, blood transfusions (due to anaemia) and splenomegaly (enlarged spleen).

The trial’s primary efficacy endpoint was TSS reduction of ≥50% over the 28 days immediately before the end of Week 24 compared to baseline TSS, using the Myelofibrosis Symptom Assessment Form. Key secondary endpoints included TI rate for ≥12 weeks immediately before the end of Week 24 with haemoglobin levels ≥ 8 g/dL and SRR based on splenic volume reduction of ≥35% at Week 24 from baseline.

Patients were randomised at 2:1 to receive either momelotinib or danazol (n=130 and n=65, respectively). After 24 weeks of treatment, patients on danazol were allowed to crossover to receive momelotinib. Early crossover to momelotinib was available for confirmed splenic progression. The trial enrolled 195 patients across 21 countries.

About momelotinib

Momelotinib is a potential new medicine with a differentiated mechanism of action, with inhibitory ability along three key signalling pathways: Janus kinase (JAK) 1, and JAK2 and activin A receptor, type I (ACVR1).1,2,3,4 Inhibition of JAK1 and JAK2 may improve constitutional symptoms and splenomegaly.1,2,4 Additionally, direct inhibition of ACVR1 leads to a decrease in circulating hepcidin, which is elevated in myelofibrosis and contributes to anaemia.1,2,3,4

Momelotinib was most recently developed by Sierra Oncology, Inc., which GSK acquired in July 2022, building on GSK’s expertise in haematology and portfolio of specialty medicines and vaccines.

About myelofibrosis

Myelofibrosis is a rare blood cancer that results from dysregulated JAK-signal transducer and activator of transcription protein signalling and is characterised by constitutional symptoms, splenomegaly, and progressive anaemia. Myelofibrosis affects approximately 20,000 patients in the US, with about 40% of patients already anaemic at the time of diagnosis and nearly all patients estimated to develop anaemia eventually.1,5 Patients will often require transfusions, and more than 30% will discontinue treatment due to anaemia.6 Anaemia and transfusion dependence strongly correlate with poor prognosis and shortened survival.7

GSK in oncology

GSK is focused on maximising patient survival through transformational medicines. GSK’s pipeline is focused on immuno-oncology, cell therapy, tumour cell targeting therapies and synthetic lethality. Our goal is to achieve a sustainable flow of new treatments based on a diversified portfolio of investigational medicines utilising modalities such as small molecules, antibodies, antibody-drug conjugates, and cell therapy, either alone or in combination.

About GSK

GSK is a global biopharma company with a purpose to unite science, technology, and talent to get ahead of disease together. Find out more at gsk.com/company

1 Chifotides, H.T., Bose, P. & Verstovsek, S. Momelotinib: an emerging treatment for myelofibrosis patients with anemia. J Hematol Oncol 15, 7 (2022). https://doi.org/10.1186/s13045-021-01157-4

2 Verstovsek S, et al. MOMENTUM: momelotinib vs danazol in patients with myelofibrosis previously treated with JAKi who are symptomatic and anemic. Future Oncol. 2021;17(12):1449-1458. https://doi.org/10.2217/fon-2020-1048

3 Asshoff M, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017;129(13):1823-1830.

4 Oh S, et al. ACVR1/JAK1/JAK2 inhibitor momelotinib reverses transfusion dependency and suppresses hepcidin in myelofibrosis phase 2 trial. Blood Adv. 2020;4(18):4282-4291.

5 Naymagon, L., & Mascarenhas, J. (2017). Myelofibrosis-Related Anemia: Current and Emerging Therapeutic Strategies. HemaSphere, 1(1), e1. https://doi.org/10.1097/HS9.0000000000000001

6 Palandri, F., Palumbo, G.A., Elli, E.M. et al. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis. Blood Cancer J. 11, 4 (2021). https://doi.org/10.1038/s41408-020-00392-1

7 Pardanani, A., & Tefferi, A. (2011). Prognostic relevance of anemia and transfusion dependency in myelodysplastic syndromes and primary myelofibrosis. Haematologica, 96(1), 8–10. https://doi.org/10.3324/haematol.2010.035519

 

Read more

What to do when GVHD attacks the gastrointestinal tract or liver

Sunday May 1, 2022

Presenter: Paul Shaughnessy MD, Sarah Cannon Transplant and Cellular Therapy Program at Methodist Hospital, San Antonio, Texas.

Presentation is 32 minutes long with 23 minutes of Q & A.

Summary: Half of patients who have a stem cell transplant using donor cells (an allogeneic transplant) develop chronic graft-versus-host-disease (GVHD). In up to a third of those patients, GVHD affects the mouth, esophagus, stomach and/or GI tract. GVHD can also affect the liver and/or pancreas. This presentation describes the symptoms and treatment options for GI and liver GVHD.

Highlights:

  • Up to a third of patients with chronic GVHD have symptoms in the esophagus, stomach and/or lower GI tract. Symptoms include stomach upset, diarrhea, difficulty swallowing, malabsorption and weight loss.
  • GVHD can damage the GI tract and enable bad bacteria to outnumber beneficial bacteria, which can lead to more GVHD and worse outcomes.
  • Jakafi® and Rezurock® have recently been approved by the FDA to treat GVHD that does not respond to steroids. Extracorproeal photopheresis (ECP) also helps some patients with GI GVHD.

Key Points:

(08:36): Use of very broad-spectrum antibiotics after transplant can destroy the good bacteria in our bowels and lead to more GVHD.

(11:46): GVHD can damage saliva glands in the mouth, causing dry mouth, mouth sores and an increased risk of cavities or periodontal disease

(13:32): Saliva substitutes can help people who have a dry mouth. If people have pain or cannot eat, Decadron® and tacrolimus rinses can help.

(17:15): GVHD can affect the pancreas, the organ that makes enzymes to helps digest food, causing fat and undigested food in the stool.

(20:39): GVHD can cause strictures in the esophagus which can make swallowing difficult. A procedure called esophageal dilation can help.

(22:21) Jaundice and an increase in bilirubin may be a sign of GVHD in the liver, which can interfere with the digestion of food.

(25:29): Mild or moderate GVHD may be treated with localized or topical agents. Severe GVHD is typically treated with systemic corticosteroids or calcineurin inhibitors.

(26:24): Jakafi®and Rezurock® have recently been approved by the FDA to treat GVHD that does not respond to steroids.

(28:14): Extracorporeal photopheresis, which is FDA-approved for patients with cutaneous T-cell lymphoma, can also help patients with GI or liver GVHD

(30:30): Several specialists including dentists, gastroenterologist, nutritionists and physical therapist may be needed to effectively treat GVHD.

Read more and view video presentation

Imago BioSciences Reports Second Quarter 2022 Financial Results and Provides Recent Business Updates

Friday, August 12, 2022 at 4:05 PM

– Positive Data from the Ongoing Phase 2 Study of Bomedemstat in Essential Thrombocythemia (ET) Presented at European Hematology Association (EHA) Meeting 2022 –

– Positive Data from the Ongoing Phase 2 Study of Bomedemstat in Advanced Myelofibrosis Presented at EHA 2022 –

– First Participant Dosed in Investigator-Sponsored Phase 1/2 Study of Bomedemstat in Combination with Atezolizumab in Small Cell Lung Cancer –

– Obtained Advice from FDA Clinical Outcomes Assessment (COA) Group with Regard to Patient Reported Outcome (PRO) Endpoints for the Company’s Planned Phase 3 Trial of Bomedemstat in ET Patients –

SOUTH SAN FRANCISCO, Calif., Aug. 12, 2022 (GLOBE NEWSWIRE) — Imago BioSciences, Inc. (“Imago” or the “Company”) (Nasdaq: IMGO), a clinical-stage biopharmaceutical company discovering and developing new medicines for the treatment of myeloproliferative neoplasms (MPNs) and other bone marrow diseases, today reported financial results for the second quarter ended June 30, 2022 and provided business updates.

“We were delighted to present updated positive data on our Phase 2 trial of bomedemstat in essential thrombocythemia (ET) at EHA in June. In our updated dataset, bomedemstat demonstrated hematologic and symptomatic improvement in ET patients and, importantly, demonstrated the durability of these hematologic responses. These data will support our ongoing discussions with the FDA, as we target an End of Phase 2 meeting with the FDA later this year, and move towards initiating the pivotal trial,” said Hugh Young Rienhoff, Jr., M.D, Chief Executive Officer of Imago.

“At EHA, we also presented positive data from our advanced myelofibrosis (MF) Phase 2 trial where bomedemstat demonstrated improvements in total symptom scores, fibrosis grades, spleen volumes and anemia, as well as reductions in mutant allele frequencies. Looking ahead, we plan to evaluate bomedemstat in combination with ruxolitinib in MF patients in a trial starting in the Fall 2022. Lastly, in addition to our long-held plans to study bomedemstat in other myeloproliferative neoplasms, such as polycythemia vera, we were pleased to see initiation of the first solid tumor clinical study of bomedemstat, an investigator-sponsored study of bomedemstat in combination with atezolizumab for small cell lung cancer.”

Read more

Study Advances Knowledge Base About Children and Adolescents With Myeloproliferative Neoplasms

August 10, 2022
Jennifer Larson

Myeloproliferative neoplasms (MPNs) are uncommon in children and young people. As a result, the guidelines currently used to reduce the risk of thrombosis and hemorrhage in patients with MPNs are based on literature involving patients over the age of 60.

A team of researchers set out to learn more about children and young adults with MPNs, due to the lack of sparse existing data. They published their findings in Blood Advances.

The researchers conducted a retrospective study that included members of the European Hematology Association MPN scientific working group who were diagnosed with an MPN before the age of 25. They examined data from 444 patients from 15 countries, 53.8% of who were diagnosed between the ages of 20 and 25.

They confirmed that the most common MPN subtype was essential thrombocythemia (ET), representing 71.6% of this cohort. Another 18.2% had the polycythemia vera (PV) subtype, and the remaining had primary myelofibrosis (PMF), prefibrotic myelofibrosis (PreMF), or unclassified MPN (MPN-u).

They also found what they called “interesting perspectives on how these MPN are perceived by physicians” and how they are managed. “Importantly, we demonstrate that standard prognostic scores do not perform well in this cohort,” they wrote. “As an example, 67.8% received at least one cytoreductive drug when only 21.4% of them should have received such therapy according to currently [European LeukemiaNet (ELN)] recommendations.”

They added that the proportion of certain prescriptions for the participants also suggested that their physicians didn’t apply ELN recommendations “where interferon is the recommended first-line therapy in patients aged below 60 years old because of its lack of leukemogenicity  and its low teratogenicity.” But as they noted, no MPN guidelines exist for children or adolescents, so the differences likely reflect “historical shifts in guidelines and availability of therapy.”

The researchers also uncovered higher-than-anticipated rates of thrombosis, hemorrhage, and transformation in their real-world cohort compared with the rates in their literature review. And 11% of patients had a history of thrombotic events, especially in patients with PV (21.5%).

“Due to the predominance of venous events, classical management with low-dose aspirin to prevent thrombosis should perhaps be questioned in this specific population, as it mainly reduces the risk of arterial events,” the researchers wrote. “The use of anticoagulants could be a way to reduce the incidence of venous thrombotic events.”

The study did have limitations, such as its retrospective nature, which may have resulted in some biases. Also, without specific treatment guidelines for young patients, the management could have been heterogeneous among the various centers, which could have influenced the rate of complications. However, they did point out that they had a “very high number of such extremely rare patients….suggesting that we revealed a reliable picture of MPN in very young patients.”

Ultimately, the research revealed a high disease burden with incidences of thrombotic events and transformations that were higher than previously reported in this particular population. The information could be used for future biomarkers studies, clinical trials, and the creation specific treatment guidelines for younger MPN patients.

Disclosures: Some authors declared affiliations with biotech, pharmaceutical, and/or device companies. Please see the original reference for a full list of authors’ disclosures.

Reference

Sobas M, Kiladjian J, Beauverd Y, et al. Real world study of children and young adults with myeloproliferative neoplasms identifying risks and unmet needsBlood Adv. Published online July 8, 2022. doi:10.1182/bloodadvances.2022007201

 

Read more