Next Article in Journal
Advanced Pancreatic Ductal Adenocarcinoma: Moving Forward
Next Article in Special Issue
Impact of Host, Lifestyle and Environmental Factors in the Pathogenesis of MPN
Previous Article in Journal
Postoperative Morbidity and Failure to Rescue in Surgery for Gastric Cancer: A Single Center Retrospective Cohort Study of 1107 Patients from 1972 to 2014
Previous Article in Special Issue
Clinical, Hematologic, Biologic and Molecular Characteristics of Patients with Myeloproliferative Neoplasms and a Chronic Myelomonocytic Leukemia-Like Phenotype
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Use of Interferon Alfa in the Treatment of Myeloproliferative Neoplasms: Perspectives and Review of the Literature

1
Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
2
Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
3
Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
*
Author to whom correspondence should be addressed.
Submission received: 26 June 2020 / Revised: 9 July 2020 / Accepted: 10 July 2020 / Published: 18 July 2020
(This article belongs to the Special Issue New Insights into Myeloproliferative Neoplasms)

Abstract

:
Interferon alfa was first used in the treatment of myeloproliferative neoplasms (MPNs) over 30 years ago. However, its initial use was hampered by its side effect profile and lack of official regulatory approval for MPN treatment. Recently, there has been renewed interest in the use of interferon in MPNs, given its potential disease-modifying effects, with associated molecular and histopathological responses. The development of pegylated formulations and, more recently, ropeginterferon alfa-2b has resulted in improved tolerability and further expansion of interferon’s use. We review the evolving clinical use of interferon in essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis (MF). We discuss interferon’s place in MPN treatment in the context of the most recent clinical trial results evaluating interferon and its pegylated formulations, and its role in special populations such as young and pregnant MPN patients. Interferon has re-emerged as an important option in MPN patients, with future studies seeking to re-establish its place in the existing treatment algorithm for MPN, and potentially expanding its use for novel indications and combination therapies.

1. Introduction

Interferons are a group of cytokines that have a host of immunomodulatory, antiproliferative, and antiangiogenic properties, and were initially named for their ability to “interfere” with viral replication [1]. Human leukocyte interferons were first purified and studied in clinical trials in the late 1970s [2]. In the 1980s, cloning and large-scale production of interferon alfa allowed its evaluation in multiple oncologic diseases [3,4], such as multiple myeloma [5], hairy cell leukemia [6], chronic myeloid leukemia (CML) [7], and Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) including polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF) [8,9]. Currently, interferon alfa is approved in the treatment of hairy cell leukemia, hepatitis B and C infections, malignant melanoma, AIDS-related Kaposi sarcoma, follicular lymphoma and condyloma acuminata, and has additional off-label use for many hematologic and solid cancers including MPNs (Table 1).
Early studies on the use of interferon alfa in MPNs demonstrated excellent hematologic responses, including normalization of platelet and leukocyte counts in ET and a reduction in phlebotomy in PV [8,9]. In vitro studies of interferon alfa treatment also showed a reduction in hematopoietic progenitor growth, with MPN clones showing greater sensitivity to interferon’s antiproliferative effects compared to normal progenitors [10,11,12]. In PV and ET patients, interferon alfa treatment was able to suppress clonal hematopoiesis, as demonstrated through early X-inactivation studies [13,14,15]. In CML, interferon represented the mainstay of treatment prior to the development of tyrosine kinase inhibitor (TKI) therapy and was the first agent able to induce sustained cytogenetic remissions [16].
However, despite its promising therapeutic properties, interferon alfa’s side effect profile and its lack of official regulatory approval were major factors in its disuse in recent decades. Only recently has interest in interferon alfa re-emerged. Reports that interferon alfa could lead to improvements in JAK2 V617F and CALR driver mutation burdens renewed further interest in its use for treatment of MPNs [17,18,19]. The development of more tolerable forms of interferon, including the pegylated form, has further expanded interest.
The purpose of this review is to discuss interferon alfa’s current role in the treatment of Philadelphia chromosome-negative MPNs, including the use of more recent formulations such as pegylated interferon and ropeginterferon, and in special populations such as young and pregnant patients.

2. Mechanism of Action of Interferon

The interferon family is categorized into three types, of which interferon alfa belongs to the Type I group and is broadly involved in the innate defense against viral infections [20]. Interferon alfa-2 has been the predominant Type I interferon used as a therapeutic agent, and includes standard interferon alfa-2b (Intron A), pegylated interferon alfa-2a (Pegasys), pegylated interferon alfa-2b (PegIntron), and, most recently, ropeginterferon alfa-2b (Besremi).
Both Type I and II interferons exert their effects through human interferon alfa receptor chains, which activate signaling via the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway [21,22]. The anti-viral properties of interferon have led to its successful use in diseases such as hepatitis B and C and Kaposi sarcoma. Interferon’s mechanism of action in MPNs is less clear. Numerous studies have demonstrated an apoptotic effect on hematopoietic progenitors, with a preference for the mutated clone [23,24]. Colony unit megakaryocyte proliferation is also directly targeted, which likely accounts for interferon’s effects on thrombocytosis. In addition, there is evidence that interferon inhibits thrombopoietin (TPO) activation via suppression of JAK2 substrate phosphorylation [25]. Mouse models have shown that interferon can directly activate dormant hematopoietic stem cells in vivo [26], and specifically can induce differentiation in JAK2 V617F mutated quiescent stem cells [27]. This results in potential long-term depletion of the mutant clone via direct targeting of progenitor cells.
However, interferon also induces broader stimulatory effects on the immune system, enhancing surveillance and targeting of the mutant clone. Studies have demonstrated that interferon augments T-cell, macrophage, and natural killer cell activity, and increases expression of tumor-associated and major histocompatibility complex antigens [28,29,30]. These immunologic changes implicate immunotherapy as a potential therapeutic modality of MPN. Additional putative mechanisms of interferon include its pro-apoptotic properties, with gene expression profiles demonstrating induction of known mediators of apoptosis such as caspase and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [31]. It is unclear how interferon’s anti-angiogenic effects contribute to its efficacy in myeloid malignancies [32], although it is possible that angiogenesis plays a role in the tumor microenvironment.

3. Clinical Use of Interferon

Essential Thrombocythemia and Polycythemia Vera

Although interferon is not currently approved for treatment in ET and PV, consensus guidelines recommend interferon as an option for first-line cytoreduction, and in particular in younger or pregnant patients (Table 1) [33,34]. Standard interferon alfa-2b has been used for decades in the treatment of ET and PV, with induction of hematologic—and in later clinical trials, molecular—remissions [8,9]. In one prospective, open-label, observational trial in 123 ET and 136 PV patients, interferon alfa demonstrated hematologic response rates of 73% and 71%, respectively [35]. This is similar to hematologic response rates with hydroxyurea [35]. Molecular responses, as defined by decreases in mutant allele burden by 20–50% (minor), greater than 50% (partial), or to undetectable levels (complete), also occurred in 55% of ET and 19% of PV patients with JAK2 V617F mutations [35]. There is no evidence that interferon can improve overall survival and alter the biologic disease course, such as reducing transformation to myelofibrosis or leukemia; however, interferon can reduce incidence of venous thromboembolism (VTE) in high-risk ET and PV patients [35,36,37].
Interferon alfa also has been reported to improve MPN-related symptoms, including a reduction in paresthesia and erythromelalgia in PV [8,9]. However, despite its clinical efficacy, interferon alfa is associated with a significant toxicity profile. Common side effects include flu-like symptoms, fever, malaise, nausea, and vomiting, with attrition rates of up to 35% reported in some clinical trials [8,9,38]. Interferon alfa is also dosed three times weekly, typically at 3 million units per dose. Given these limitations, pegylated forms of interferon (pegylated interferon alfa-2a and 2b) have been developed, which has improved tolerability and can be given once a week. Clinical trials of over 400 ET and PV patients have demonstrated overall hematologic remission rates of approximately 80% with pegylated interferon, including freedom from phlebotomy in 60% of PV patients [8,9]. Pegylated interferon is also associated with molecular response rates in 20–60% of JAK2-mutated patients [18]. Interestingly, the presence of concomitant non-driver mutations is associated with smaller mean decreases in JAK2 V617F allele burden with pegylated interferon treatment [39]. Interferon may have decreased ability to eradicate TET2-positive clones even with elimination of the JAK2 V617F-mutant clone [10], suggesting that the genomic landscape of MPN patients can predict responses to interferon treatment. Similarly, pegylated interferon alfa can induce hematologic and molecular responses in CALR-mutated ET patients [40]. In a cohort of 31 CALR-mutated ET patients, hematologic responses were achieved in all patients and median CALR variant allele fraction decreased from 41% to 26%, with two patients achieving complete molecular remission. Decreases in CALR allele fraction correlate closely with other measures of disease burden including lactate dehydrogenase, hemoglobin, and white blood cell and platelet counts [41]. Similar to what has been found in JAK2 V617F-mutated patients, the presence of additional mutations such as TET2, ASXL1, IDH2, and TP53 was associated with poorer molecular responses [40]. There is evidence that molecular responses are more easily obtained in JAK2 V617F compared to CALR patients, thought to be due to increased interferon sensitivity of JAK2 V617F-mutant clones [42].
These responses are also durable, which is an appealing aspect of interferon treatment. In a long-term follow up of 83 ET and PV patients treated with pegylated interferon, median duration of hematologic and molecular response was 66 and 53 months, respectively [43]. In addition, three patients were able to sustain a complete molecular remission even after discontinuation of therapy, although in most patients JAK2 V617 allele burden eventually increased after the first 24 months of treatment [43]. However, disease evolution remains similar to historical data in patients treated with other therapies [44], with a reported 8% transformation rate to myelofibrosis or leukemia [43]. VTE, the most common disease complication in ET and PV, occurred at a rate of 1.22 per 100 person-years, and is decreased compared to untreated high-risk ET and PV patients [43,45,46].
Recently, results from randomized trials comparing pegylated interferon head-to-head with hydroxyurea have been reported. MPN-RC-112 was a Phase III trial that randomized high-risk ET/PV patients to first-line treatment with either pegylated interferon alfa-2a or hydroxyurea (Table 2) [47,48]. The overall response rate of 78% with pegylated interferon was not significantly different from the overall response rate of 70% in hydroxyurea-treated patients. Best bone marrow responses occurred in 17% of evaluable patients treated with pegylated interferon, compared with 33% of patients treated with hydroxyurea (p = 0.052). Similarly, the Phase III DALIAH trial randomized 205 previously untreated MPN patients to interferon alfa-2a, interferon alfa-2b, or hydroxyurea, and interim analysis observed a 49% overall response rate in ET, PV, and pre-MF patients treated with interferon alfa, although this was significantly lower than the 75% response rate seen with hydroxyurea [49]. However, as pegylated interferon-treated patients often have continued clinical improvements over time, we await final analysis results with longer follow up. Partial molecular responses occurred in 16% of interferon-treated patients, compared to 23% in hydroxyurea-treated patients, with one complete molecular remission.
Pegylated interferon has also been studied specifically in ET and PV patients previously treated with hydroxyurea, a population that is associated with worse outcomes (Table 2) [51]. The Phase II single-arm MPD-RC-111 study evaluated pegylated interferon in 65 ET and 50 PV high-risk patients who were refractory to or intolerant of hydroxyurea [52]. Overall response rates at 12 months were 69% and 60%, respectively, in ET and PV patients, with improved responses seen in CALR-mutated patients. Complete response rates of 43% and 22% in ET and PV were lower compared to other studies, which can be attributed to the high-risk features of this study population. Bone marrow and molecular remissions were rare although follow up was shorter, with a median of 19.6 months. Arterial and venous thrombosis occurred in 2% and 5% of patients at 1 and 2 years, respectively, and 1 PV patient in the cohort progressed to myelofibrosis.
Although pegylated interferon has improved tolerability compared to standard interferon, nearly all patients experience some adverse effects, with the most common being fatigue, myalgia, and nausea or vomiting and diarrhea. Neuropsychiatric side effects such as depression are also frequently encountered, with grade 3 symptoms occurring in 15% of ET and PV patients treated with pegylated interferon in one clinical trial [43]. Therefore, neuropsychiatric history should be considered prior to starting interferon, given the possibility of worsening symptoms. In addition, although toxicities improve with time, new late adverse effects including fatigue and depression occurred in 10–17% of patients annually after 24 months [43]. Laboratory adverse effects of pegylated interferon include hematologic abnormalities such as leukopenia, anemia, and thrombocytopenia, as well as elevation of liver function tests. Late autoimmune toxicities such as hypothyroidism, vasculitis, or hepatitis can also occur. When compared to hydroxyurea in MPD-RC-112 and DALIAH, grade 3/4 adverse events were significantly higher in pegylated interferon [47,49]. Discontinuation rates of pegylated interferon are still typically less than that of standard interferon, occurring in approximately 15–20% of patients on clinical trial [43,47,52], although in DALIAH, toxicity-dependent discontinuation was significantly increased in pegylated interferon (27%) compared to hydroxyurea (5%) [49]. Given that fatigue is also a common symptom in MPN patients, the non-trivial side effect profile of pegylated interferon makes it a less ideal choice for first-line treatment in patients who have significant disease-related symptom burden.
Further efforts to improve interferon’s tolerability and administration have resulted in the recent development of ropeginterferon alfa-2b, which is a monopegylated interferon alfa given subcutaneously. Ropeginterferon’s extended half-life allows every other week dosing with extension to a monthly maintenance schedule. Its formulation as a home self-administered prefilled pen further improves patient compliance compared to pegylated interferon alfa-2a, the latter of which may pose unique challenges to patients such as dispensation of very small, difficult-to-measure volumes. A few recent trials have reported on the efficacy of ropeginterferon in PV patients (Table 3). The non-inferiority randomized Phase III PROUD-PV trial and its extension study CONTINUATION-PV evaluated ropeginterferon versus hydroxyurea in 257 PV patients with fewer than 3 years of prior cytoreductive treatment [53]. The primary endpoint was a composite outcome of hematologic response and spleen volume reduction. Given that few PV patients had enlarged spleens at enrollment, the trial failed to demonstrate the non-inferiority of ropeginterferon at 12 months. However, ropeginterferon was effective at inducing hematologic response, with 43% of patients experiencing complete hematologic response. The complete hematologic response rate in CONTINUATION-PV was significantly improved in ropeginterferon-treated patients compared to hydroxyurea-treated patients, and responses to ropeginterferon continued to increase over time. Molecular responses were not different between groups at 1 year (34% versus 42%), but were significantly improved in CONTINUATION-PV at 3 years (68% versus 33%). In addition to JAK2 V617F, ropeginterferon was also shown to reduce TET2 allele burden, a mutation associated with inflammation that may contribute to disease acceleration in MPNs [53,54,55]. Ropeginterferon is also being evaluated in the low-risk PV population specifically, with interim analysis results of 100 patients recently reported for the LOW-PV study, a Phase II randomized trial comparing ropeginterferon with phlebotomy alone [56]. A total of 84% of ropeginterferon-treated patients met the composite endpoint of maintaining a hematocrit ≤ 45% for 12 months in the absence of progressive disease, compared to 60% of patients in the standard arm (p = 0.008), allowing the trial to stop early due to efficacy. This supports the use of ropeginterferon in the low-risk PV population as a means to reduce phlebotomy and maintain target hematocrit goals.
Similarly, the Phase I/II study PEGINVERA evaluated efficacy and safety of ropeginterferon alfa in 51 PV patients regardless of prior cytoreductive therapy [57]. Patients who responded well following 1 year of biweekly treatment were able to switch to a more convenient monthly administration with maintenance scheduling, which occurred at a median of 2 years. Preliminary results reported that nearly all patients (41/42) had a hematologic response, with complete responses occurring in 64% of patients after a median of 34 weeks. Complete and partial molecular responses also occurred in 28.6% and 45.2% of all patients. Significantly, there was an over 80% complete hematologic and molecular response rate in patients who were able to switch over to monthly maintenance dosing, with 100% compliance.
The most common adverse events reported with ropeginterferon include thrombocytopenia, leukopenia, increased liver function tests, arthralgia, flu-like symptoms and fatigue. The majority of toxicities were mild, and discontinuation rates in PROUD-PV and CONTINUATION-PV were 8% [53,58]. PEGINVERA utilized a maximum-tolerated dose design during the first year, resulting in discontinuation of ropeginterferon in 21 out of the 51 patients [57]. As only 8 of the 21 patients discontinued after the first year due to treatment-related adverse effects, the investigators recommended slower uptitration of ropeginterferon for better tolerability. LOW-PV also reported 6% grade ≥3 adverse events in ropeginterferon-treated patients, which was not significantly higher than standard therapy patients, again indicating its potential role in the low-risk PV population [56].
Given these favorable results, the Committee for Medicinal Products for Human Use (CHMP) has granted marketing authorization for ropeginterferon in Europe, and the Food and Drug Administration (FDA) has accepted a biologics license application for ropeginterferon’s approval in the United States. If approved, ropeginterferon would be the first interferon developed specifically for MPN. Ropeginterferon is also currently being evaluated in essential thrombocythemia (NCT 04285086).

4. Myelofibrosis

Interferon’s direct effects on clonal hematopoietic stem cells have generated interest in its use as a potential disease-modifying agent in MF. Several studies have demonstrated that both standard and pegylated interferon result in significantly decreased bone marrow fibrosis in a subset of MF patients with clinical response [58,59]. However, overall disease response has been disappointing [8,9]. In addition, toxicity, particularly hematologic toxicity with dose-limiting cytopenias, are more common in advanced MF compared to PV and ET, leading to more frequent discontinuation [8,9]. To date, allogeneic hematopoietic stem cell transplantation is the only known treatment that can modify the disease course in MF.
Studies evaluating interferon use in lower-risk MF patients have been more encouraging. A retrospective study of 62 MF patients treated with pegylated interferon demonstrated splenic response in 47%, constitutional symptom improvement in 82%, and resolution of leukocytosis and thrombocytosis in 60–80% of patients [60]. The majority of patients were low or intermediate risk, with only 21.3% of patients being high risk by International Prognostic Scoring System (IPSS). A prospective study of 30 Dynamic IPSS (DIPSS) low- or intermediate-1-risk MF patients demonstrated an overall response rate of 50% (including complete and partial remissions as well as clinical improvement) [61]. Forty percent of patients experienced a ≥50% decrease in spleen size, and of the 25 patients with available bone marrow biopsies, 5 showed a reduction in reticulin fibrosis. Patients without high-molecular-risk (HMR) mutations, including ASXL1, had significantly better responses. Interferon is recommended as an option in symptomatic low- or intermediate-risk MF patients by the National Comprehensive Cancer Network, although European Leukemia Net recommends consideration of hydroxyurea or ruxolitinib as first-line treatment in lower-risk MF patients requiring cytoreduction [33,62]. Ultimately, the decision for which cytoreductive agent to use in these patients is based on individual factors such as comorbidities, risk status, symptom burden, age, and toxicity profile.

5. Combination Therapy

While interferons can induce remissions, not all patients respond, and side effects are frequently limiting. Combination therapy of interferon with JAK2 inhibitors such as ruxolitinib can potentially enhance interferon signaling, given that high levels of inflammation may be involved with resistance to interferon, and JAK2 inhibitors have anti-inflammatory properties [63]. Combination therapy may also permit lower doses, thus improving tolerability. The Phase II COMBI clinical trial evaluated ruxolitinib and pegylated interferon alfa-2a in 32 PV and 18 MF patients, of which 46 patients were intolerant of or refractory to pegylated interferon monotherapy [64]. There were no high-risk MF patients included in this trial, with the majority being low or intermediate-1 risk by DIPSS+. PV patients were included if they had evidence of active disease with need for cytoreduction. Thirty-one percent and forty-four percent of PV and MF patients achieved remission. The discontinuation rates were 6% for PV and 32% for MF patients, and overall symptom scores, as measured by MPN Symptom Assessment Forms (SAFs), decreased from 22 to 15. There was a significant decrease in JAK2 V617F allele burden at 2 years. Similarly, the phase I/II RUXOPEG trial evaluated ruxolitinib and pegylated interferon alfa-2a in treatment-naïve IPSS intermediate- or high-risk MF patients [65]. Investigators found significant decreases in spleen size and JAK2 V617F allele burden, as well as improvement in hematologic parameters in evaluable patients. These data suggest that combination therapy with ruxolitinib and interferon may prove to be an effective treatment with acceptable toxicity. Table 4 provides a summary of clinical trials evaluating pegylated interferon with ruxolitinib.

6. Special Populations

6.1. Young Populations

Hydroxyurea is typically used as first-line cytoreduction in MPN patients. Although no difference has been found in secondary cancer rates between interferon and hydroxyurea [66], there are still concerns for hydroxyurea’s long-term effects [44,67,68]. Interferon’s potential for durable responses and disease modification also makes it a more appealing choice for younger patients, given that treatment can last for decades. Currently, expert consensus guidelines recommend consideration of interferon for first-line treatment in patients who are younger or who defer hydroxyurea [33,34].

6.2. Pregnant Populations

Although MPNs are typically a disease of older populations, up to 20% of patients are diagnosed before the age of 40 [69]. This introduces the specialized issue of pregnancy, with the majority of cases occurring in ET and very few occurring in MF. Pregnancy in MPNs is not contraindicated, with overall good outcomes and live birth rates of 70% in ET and PV [70,71]. However, MPN patients are at increased risk for pregnancy related complications, the most common being spontaneous abortion. Other common pregnancy related complications include higher risk for thrombosis and hemorrhage, pre-eclampsia, and stillbirths [70,72,73]. Cytoreduction is considered in patients who have higher risk features following guidelines for cytoreduction in MPN and may be given in those with prior pregnancy losses and/or complications, history of thrombosis, or extreme thrombocytosis leading to increased risk of bleeding [33,70,74].
The most commonly prescribed drug for cytoreduction in ET and PV patients, hydroxyurea, is a Category D drug in pregnancy, meaning that evidence of human risk to the fetus exists. Ruxolitinib is also contraindicated in pregnancy. Although interferons are a Category C drug, in which no controlled studies have been conducted in animals or humans, in general the literature has demonstrated safety of interferon for use in pregnancy [70,75]. Pharmacologic studies have not detected interferon in fetal blood or amniotic fluid [76]. Of 78 pregnancies treated with interferon reported in the literature, the live birth rate was 94%, with no fetal malformations [70]. Indeed, a meta-analysis showed improved live birth rates in ET women who were treated with interferon during their pregnancy [73]. Despite the association with improved outcomes, interferon use is typically reserved for high-risk pregnancies and it is unknown whether there would be any benefit in patients without traditional indication for cytoreduction.
There is less experience with pegylated interferon in pregnancy, although placental transfer of pegylated interferon is thought to be unlikely. Given its improved tolerability, it is reasonable for use instead of standard interferon in pregnant MPN patients. A case series of 10 ET pregnancies in 8 women starting treatment before conception with pegylated interferon reported 9 live births and 1 miscarriage, with no major bleeding or thrombosis events [77]. Current expert consensus guidelines recommend either interferon or pegylated interferon as first-line treatment when cytoreduction is needed in patients who are pregnant or considering pregnancy [33].
Interferons are present in breastmilk. However, the amounts are very low and it is unlikely that it will pose a significant risk in nursing mothers and breastfeeding infants [78]. Therefore, breastfeeding can be considered in women receiving interferon after delivery.

7. Conclusions

Despite being a decades-old drug, interferon remains one of the only treatments with the potential for disease modification, given its impact on mutation burdens and induction of durable responses. However, its administration and toxicity profile were major barriers to its widespread use, resulting in the development of pegylated interferon and, more recently, ropeginterferon. Large meta-analyses of interferon’s effects are difficult due to different drug formulations and endpoints, but, overall, clinical trials have demonstrated clinical responses in the vast majority of ET and PV patients, as well as some molecular responses. Pegylated formulations have overall shown similar efficacy to hydroxyurea, and recent encouraging data surrounding ropeginterferon have led to ongoing efforts for official regulatory approval in MPNs. As a result, interferons are a reasonable cytoreductive alternative to hydroxyurea in ET and PV, and are recommended for first-line consideration in younger patients and pregnant women. Although interferon has limited efficacy in advanced MF, it does have use in lower-risk MF patients, with ability to improve symptoms, spleen size, and laboratory parameters. There is ongoing investigation on combination therapies with pegylated interferon and ruxolitinib in order to further improve efficacy and tolerability. The renewed interest in interferon will undoubtedly re-establish its place as an important component in the treatment of MPNs.

Author Contributions

J.H. and G.H. designed, wrote, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

J.H. has no conflicts of interest to report. G.H. receives grant support from the K-12 Paul Calabresi award, the ASH-AMFDP award and the Sanchez Ferguson award; research support from Bayer, Incyte and Merck; and consulting support from Incyte, Celgene, Agios and Jazz pharmaceuticals.

References

  1. Isaacs, A.; Lindenmann, J. Virus interference. I. The interferon. Proc. R. Soc.Lond. Ser. B-Biol. Sci. 1957, 147, 258–267. [Google Scholar]
  2. Cantell, K.; Hirvonen, S.; Kauppinen, H.-L.; Myllylä, G. [4] Production of interferon in human leukocytes from normal donors with the use of Sendai virus. Methods Enzymol. 1981, 78, 29–38. [Google Scholar] [CrossRef] [PubMed]
  3. Derynck, R.; Content, J.; Declercq, E.; Volckaert, G.; Tavernier, J.; Devos, R.; Fiers, W. Isolation and structure of a human fibroblast interferon gene. Nature 1980, 285, 542–547. [Google Scholar] [CrossRef] [PubMed]
  4. Maeda, S.; McCandliss, R.; Gross, M.; Sloma, A.; Familletti, P.C.; Tabor, J.M.; Evinger, M.; Levy, W.P.; Pestka, S. Construction and identification of bacterial plasmids containing nucleotide sequence for human leukocyte interferon. Proc. Natl. Acad. Sci. USA 1980, 77, 7010–7013. [Google Scholar] [CrossRef] [Green Version]
  5. Åhre, A.; Björkholm, M.; Österborg, A.; Brenning, G.; Gahrton, G.; Gyllenhammar, H.; Holm, G.; Johansson, B.; Juliusson, G.; Järnmark, M.; et al. High doses of natural α-interferon (α-IFN) in the treatment of multiple myeloma—A pilot study from the Myeloma Group of Central Sweden (MGCS). Eur. J. Haematol. 2009, 41, 123–130. [Google Scholar] [CrossRef] [PubMed]
  6. Habermann, T.M.; Andersen, J.W.; Cassileth, P.A.; Bennett, J.M.; Oken, M.M. Sequential administration of recombinant interferon alpha and deoxycoformycin in the treatment of hairy cell leukaemia. Br. J. Haematol. 1992, 80, 466–471. [Google Scholar] [CrossRef]
  7. Hehlmann, R.; Heimpel, H.; Hasford, J.; Kolb, H.J.; Pralle, H.; Hossfeld, D.K.; Queisser, W.; Loffler, H.; Hochhaus, A.; Heinze, B.; et al. Randomized comparison of interferon-alpha with busulfan and hydroxyurea in chronic myelogenous leukemia. The German CML Study Group. Blood 1994, 84, 4064–4077. [Google Scholar] [CrossRef] [Green Version]
  8. Kiladjian, J.-J.; Chomienne, C.; Fenaux, P. Interferon-α therapy in bcr-abl-negative myeloproliferative neoplasms. Leukemia 2008, 22, 1990–1998. [Google Scholar] [CrossRef] [Green Version]
  9. Kiladjian, J.-J.; Mesa, R.A.; Hoffman, R. The renaissance of interferon therapy for the treatment of myeloid malignancies. Blood 2011, 117, 4706–4715. [Google Scholar] [CrossRef] [Green Version]
  10. Kiladjian, J.-J.; Masse, A.; Cassinat, B.; Mokrani, H.; Teyssandier, I.; Le Couédic, J.-P.; Cambier, N.; Almire, C.; Pronier, E.; Casadevall, N.; et al. Clonal analysis of erythroid progenitors suggests that pegylated interferon α-2a treatment targets JAK2V617F clones without affecting TET2 mutant cells. Leukemia 2010, 24, 1519–1523. [Google Scholar] [CrossRef]
  11. Castello, G.; Lerza, R.; Cerruti, A.; Cavallini, D.; Bogliolo, G.; Pannacciulli, I. The in vitro and in vivo effect of recombinant interferon α-2a on circulating haemopoietic progenitors in polycythaemia vera. Br. J. Haematol. 1994, 87, 621–623. [Google Scholar] [CrossRef] [PubMed]
  12. Carlo-Stella, C.; Cazzola, M.; Gasner, A.; Barosi, G.; Dezza, L.; Meloni, F.; Pedrazzoli, P.; Hoelzer, D.; Ascari, E. Effects of recombinant alpha and gamma interferons on the in vitro growth of circulating hematopoietic progenitor cells (CFU-GEMM, CFU-Mk, BFU-E, and CFU-GM) from patients with myelofibrosis with myeloid metaplasia. Blood 1987, 70, 1014–1019. [Google Scholar] [CrossRef] [PubMed]
  13. Hino, M.; Futami, E.; Okuno, S.; Miki, T.; Nishizawa, Y.; Morii, H. Possible selective effects of Interferon α-2b on a malignant clone in a case of polycythemia vera. Ann. Hematol. 1993, 66, 161–162. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, E.; Jelinek, J.; Pastore, Y.D.; Guan, Y.; Prchal, J.F.; Prchal, J.T. Discrimination of polycythemias and thrombocytoses by novel, simple, accurate clonality assays and comparison with PRV-1 expression and BFU-E response to erythropoietin. Blood 2003, 101, 3294–3301. [Google Scholar] [CrossRef]
  15. Massaro, P.; Foa, P.; Pomati, M.; LaTargia, M.L.; Iurlo, A.; Clerici, C.; Caldiera, S.; Fornier, M.; Maiolo, A.T. Polycythemia vera treated with recombinant interferon-alpha 2a: Evidence of a selective effect on the malignant clone. Am. J. Hematol. 1997, 56, 126–128. [Google Scholar] [CrossRef]
  16. Italian Cooperative Study Group on Chronic Myeloid Leukemia; Tura, S.; Baccarani, M.; Zuffa, E.; Russo, D.; Fanin, R.; Zaccaria, A.; Fiacchini, M. Interferon Alfa-2a as Compared with Conventional Chemotherapy for the Treatment of Chronic Myeloid Leukemia. New Engl. J. Med. 1994, 330, 820–825. [Google Scholar] [CrossRef]
  17. Kiladjian, J.-J.; Cassinat, B.; Turlure, P.; Cambier, N.; Roussel, M.; Bellucci, S.; Menot, M.-L.; Massonnet, G.; Dutel, J.-L.; Ghomari, K.; et al. High molecular response rate of polycythemia vera patients treated with pegylated interferon -2a. Blood 2006, 108, 2037–2040. [Google Scholar] [CrossRef]
  18. Quintás-Cardama, A.; Kantarjian, H.; Manshouri, T.; Luthra, R.; Estrov, Z.; Pierce, S.; Richie, M.A.; Borthakur, G.; Konopleva, M.; Cortes, J.; et al. Pegylated Interferon Alfa-2a Yields High Rates of Hematologic and Molecular Response in Patients With Advanced Essential Thrombocythemia and Polycythemia Vera. J. Clin. Oncol. 2009, 27, 5418–5424. [Google Scholar] [CrossRef] [Green Version]
  19. Cassinat, B.; Verger, E.; Kiladjian, J.-J. Interferon Alfa Therapy in CALR-Mutated Essential Thrombocythemia. N. Engl. J. Med. 2014, 371, 188–189. [Google Scholar] [CrossRef]
  20. Pestka, S.; Krause, C.D.; Walter, M. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef]
  21. Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef] [PubMed]
  22. Trinchieri, G. Type I interferon: Friend or foe? J. Exp. Med. 2010, 207, 2053–2063. [Google Scholar] [CrossRef] [PubMed]
  23. Lu, M.; Zhang, W.; Li, Y.; Berenzon, D.; Wang, X.; Wang, J.; Mascarenhas, J.; Xu, M.; Hoffman, R. Interferon-α targets JAK2V617F-positive hematopoietic progenitor cells and acts through the p38 MAPK pathway. Exp. Hematol. 2010, 38, 472–480. [Google Scholar] [CrossRef] [Green Version]
  24. Raefsky, E.L.; Platanias, L.C.; Zoumbos, N.C.; Young, N.S. Studies of interferon as a regulator of hematopoietic cell proliferation. J. Immunol. 1985, 135, 2507–2512. [Google Scholar] [PubMed]
  25. Wang, Q.; Miyakawa, Y.; Fox, N.; Kaushansky, K. Interferon-alpha directly represses megakaryopoiesis by inhibiting thrombopoietin-induced signaling through induction of SOCS-1. Blood 2000, 96, 2093–2099. [Google Scholar] [CrossRef] [PubMed]
  26. Essers, M.A.G.; Offner, S.; Blanco-Bose, W.E.; Waibler, Z.; Kalinke, U.; Duchosal, M.A.; Trumpp, A. IFNα activates dormant haematopoietic stem cells in vivo. Nature 2009, 458, 904–908. [Google Scholar] [CrossRef] [PubMed]
  27. Mullally, A.; Bruedigam, C.; Poveromo, L.; Heidel, F.H.; Purdon, A.; Vu, T.; Austin, R.; Heckl, D.; Breyfogle, L.J.; Kuhn, C.P.; et al. Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-α in a murine model of polycythemia vera. Blood 2013, 121, 3692–3702. [Google Scholar] [CrossRef] [Green Version]
  28. Tamura, T.; Yanai, H.; Savitsky, D.; Taniguchi, T. The IRF Family Transcription Factors in Immunity and Oncogenesis. Annu. Rev. Immunol. 2008, 26, 535–584. [Google Scholar] [CrossRef]
  29. Paquette, R.L.; Hsu, N.; Said, J.; Mohammed, M.; Rao, N.P.; Shih, G.; Schiller, G.; Sawyers, C.; A Glaspy, J. Interferon-α induces dendritic cell differentiation of CML mononuclear cells in vitro and in vivo. Leukemia 2002, 16, 1484–1489. [Google Scholar] [CrossRef]
  30. Chen, X.; Regn, S.; Raffegerst, S.; Kolb, H.J.; Roskrow, M. Interferon alpha in combination with GM-CSF induces the differentiation of leukaemic antigen-presenting cells that have the capacity to stimulate a specific anti-leukaemic cytotoxic T-cell response from patients with chronic myeloid leukaemia. Br. J. Haematol. 2000, 111, 596–607. [Google Scholar] [CrossRef]
  31. Chawla-Sarkar, M.; Lindner, D.J.; Liu, Y.-F.; Williams, B.R.; Sen, G.C.; Silverman, R.H.; Borden, E.C. Apoptosis and interferons: Role of interferon-stimulated genes as mediators of apoptosis. Apoptosis 2003, 8, 237–249. [Google Scholar] [CrossRef] [PubMed]
  32. Bonanno, L. Interferon-α as angiogenesis inhibitor: Learning from tumor models. Autoimmunity 2010, 43, 244–247. [Google Scholar] [CrossRef]
  33. Network, N.C.C. Myeloproliferative Neoplasms (Version 2.2019). Available online: https://www.nccn.org/professionals/physician_gls/pdf/mpn_blocks.pdf (accessed on 17 August 2019).
  34. Barbui, T.; Tefferi, A.; Vannucchi, A.M.; Passamonti, F.; Silver, R.T.; Hoffman, R.; Verstovsek, S.; Mesa, R.; Kiladjian, J.-J.; Hehlmann, R.; et al. Philadelphia chromosome-negative classical myeloproliferative neoplasms: Revised management recommendations from European LeukemiaNet. Leukemia 2018, 32, 1057–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, B.-T.; Zeng, Q.-C.; Zhao, W.-H.; Li, B.-S.; Chen, R.-L. Interferon α-2b gains high sustained response therapy for advanced essential thrombocythemia and polycythemia vera with JAK2V617F positive mutation. Leuk. Res. 2014, 38, 1177–1183. [Google Scholar] [CrossRef] [PubMed]
  36. Radin, A.I.; Kim, H.T.; Grant, B.W.; Bennett, J.M.; Kirkwood, J.M.; Stewart, J.A.; Hahn, R.G.; Dutcher, J.P.; Wiernik, P.H.; Oken, M.M. Phase II study of ?2 interferon in the treatment of the chronic myeloproliferative disorders (E5487). Cancer 2003, 98, 100–109. [Google Scholar] [CrossRef] [PubMed]
  37. Saba, R.; Jabbour, E.; Giles, F.; Cortes, J.; Talpaz, M.; O’Brien, S.; Freireich, E.J.; Garcia-Manero, G.; Kantarjian, H.; Verstovsek, S. Interferon α therapy for patients with essential thrombocythemia. Cancer 2005, 103, 2551–2557. [Google Scholar] [CrossRef]
  38. Quesada, J.R.; Talpaz, M.; Rios, A.; Kurzrock, R.; Gutterman, J.U. Clinical toxicity of interferons in cancer patients: A review. J. Clin. Oncol. 1986, 4, 234–243. [Google Scholar] [CrossRef]
  39. Quintás-Cardama, A.; Abdel-Wahab, O.; Manshouri, T.; Kilpivaara, O.; Cortes, J.; Roupie, A.L.; Zhang, S.-J.; Harris, D.; Estrov, Z.; Kantarjian, H.; et al. Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon α-2a. Blood 2013, 122, 893–901. [Google Scholar] [CrossRef] [Green Version]
  40. Verger, E.; Cassinat, B.; Chauveau, A.; Dosquet, C.; Giraudier, S.; Schlageter, M.-H.; Ianotto, J.-C.; Yassin, M.A.; Al-Dewik, N.; Carillo, S.; et al. Clinical and molecular response to interferon-α therapy in essential thrombocythemia patients with CALR mutations. Blood 2015, 126, 2585–2591. [Google Scholar] [CrossRef] [Green Version]
  41. Kjær, L.; Cordua, S.; Holmström, M.O.; Thomassen, M.; A Kruse, T.; Pallisgaard, N.; Larsen, T.S.; De Stricker, K.; Skov, V.; Hasselbalch, H.C. Differential Dynamics of CALR Mutant Allele Burden in Myeloproliferative Neoplasms during Interferon Alfa Treatment. PLoS ONE 2016, 11, e0165336. [Google Scholar] [CrossRef] [Green Version]
  42. Czech, J.; Cordua, S.; Weinbergerova, B.; Baumeister, J.; Crepcia, A.; Han, L.; Maié, T.; Costa, I.G.; Denecke, B.; Maurer, A.; et al. JAK2V617F but not CALR mutations confer increased molecular responses to interferon-α via JAK1/STAT1 activation. Leukemia 2018, 33, 995–1010. [Google Scholar] [CrossRef] [PubMed]
  43. Masarova, L.; Patel, K.P.; Newberry, K.J.; Cortes, J.; Borthakur, G.; Konopleva, M.; Estrov, Z.; Kantarjian, H.; Verstovsek, S. Pegylated interferon alfa-2a in patients with essential thrombocythaemia or polycythaemia vera: A post-hoc, median 83 month follow-up of an open-label, phase 2 trial. Lancet Haematol. 2017, 4, e165–e175. [Google Scholar] [CrossRef] [Green Version]
  44. Kiladjian, J.-J.; Rain, J.-D.; Bernard, J.-F.; Briére, J.; Chomienne, C.; Fenaux, P. Long-Term Incidence of Hematological Evolution in Three French Prospective Studies of Hydroxyurea and Pipobroman in Polycythemia Vera and Essential Thrombocythemia. Semin. Thromb. Hemost. 2006, 32, 417–421. [Google Scholar] [CrossRef] [PubMed]
  45. Marchioli, R.; Finazzi, G.; Landolfi, R.; Kutti, J.; Gisslinger, H.; Patrono, C.; Marilus, R.; Villegas, A.; Tognoni, G.; Barbui, T. Vascular and Neoplastic Risk in a Large Cohort of Patients With Polycythemia Vera. J. Clin. Oncol. 2005, 23, 2224–2232. [Google Scholar] [CrossRef]
  46. Barbui, T.; Vannucchi, A.M.; Buxhofer-Ausch, V.; De Stefano, V.; Betti, S.; Rambaldi, A.; Rumi, E.; Ruggeri, M.; Rodeghiero, F.; Randi, M.L.; et al. Practice-relevant revision of IPSET-thrombosis based on 1019 patients with WHO-defined essential thrombocythemia. Blood Cancer J. 2015, 5, e369. [Google Scholar] [CrossRef]
  47. Mascarenhas, J.; Kosiorek, H.E.; Prchal, J.T.; Rambaldi, A.; Berenzon, D.; Yacoub, A.; Harrison, C.N.; McMullin, M.F.; Vannucchi, A.M.; Ewing, J.; et al. Results of the Myeloproliferative Neoplasms—Research Consortium (MPN-RC) 112 Randomized Trial of Pegylated Interferon Alfa-2a (PEG) Versus Hydroxyurea (HU) Therapy for the Treatment of High Risk Polycythemia Vera (PV) and High Risk Essential Thrombocythemia (ET). Blood 2018, 132, 577. [Google Scholar] [CrossRef]
  48. Yacoub, A.; Mascarenhas, J.; Mesa, R.A.; Kosiorek, H.E.; Rampal, R.K.; Silver, R.T.; Salama, M.E.; Siwoski, O.; Dueck, A.C.; Sandy, L.; et al. Final Results of Prospective Treatment with Pegylated Interferon Alfa-2a for Patients with Polycythemia Vera and Essential Thrombocythemia in First and Second-Line Settings. Blood 2019, 134, 2943. [Google Scholar] [CrossRef]
  49. Knudsen, T.A.; Hansen, D.L.; Ocias, L.F.; Bjerrum, O.W.; Brabrand, M.; El Fassi, D.; Frederiksen, M.; Kjær, L.; Kristensen, T.K.; A Kruse, T.; et al. Long-Term Efficacy and Safety of Recombinant Interferon Alpha-2 Vs. Hydroxyurea in Polycythemia Vera: Preliminary Results from the Three-Year Analysis of the Daliah Trial-a Randomized Controlled Phase III Clinical Trial. Blood 2018, 132, 580. [Google Scholar] [CrossRef]
  50. Mascarenhas, J.O.; Prchal, J.T.; Rambaldi, A.; Mesa, R.A.; Berenzon, D.; Yacoub, A.; Harrison, C.N.; McMullin, M.F.; Vannucchi, A.M.; Ewing, J.C.; et al. Interim Analysis of the Myeloproliferative Disorders Research Consortium (MPD-RC) 112 Global Phase III Trial of Front Line Pegylated Interferon Alpha-2a Vs. Hydroxyurea in High Risk Polycythemia Vera and Essential Thrombocythemia. Blood 2016, 128, 479. [Google Scholar] [CrossRef]
  51. Alvarez-Larrán, A.; Pereira, A.; Cervantes, F.; Arellano-Rodrigo, E.; Hernández-Boluda, J.C.; Ferrer-Marín, F.; Angona, A.; Gómez, M.; Muiña, B.; Guillén, H.; et al. Assessment and prognostic value of the European LeukemiaNet criteria for clinicohematologic response, resistance, and intolerance to hydroxyurea in polycythemia vera. Blood 2012, 119, 1363–1369. [Google Scholar] [CrossRef] [Green Version]
  52. Yacoub, A.; Mascarenhas, J.O.; Kosiorek, H.; Prchal, J.T.; Berenzon, D.; Baer, M.R.; Ritchie, E.; Silver, R.T.; Kessler, C.; Winton, E.; et al. Pegylated interferon alfa-2a for polycythemia vera or essential thrombocythemia resistant or intolerant to hydroxyurea. Blood 2019, 134, 1498–1509. [Google Scholar] [CrossRef] [PubMed]
  53. Gisslinger, H.; Klade, C.; Georgiev, P.; Krochmalczyk, D.; Gercheva-Kyuchukova, L.; Egyed, M.; Rossiev, V.; Dulicek, P.; Illes, A.; Pylypenko, H.; et al. Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): A randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol. 2020, 7, e196–e208. [Google Scholar] [CrossRef]
  54. Jaiswal, S.; Natarajan, P.; Silver, A.J.; Gibson, C.J.; Bick, A.; Shvartz, E.; McConkey, M.; Gupta, N.; Gabriel, S.; Ardissino, D.; et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. New Engl. J. Med. 2017, 377, 111–121. [Google Scholar] [CrossRef]
  55. Kameda, T.; Shide, K.; Yamaji, T.; Kamiunten, A.; Sekine, M.; Taniguchi, Y.; Hidaka, T.; Kubuki, Y.; Shimoda, H.; Marutsuka, K.; et al. Loss of TET2 has dual roles in murine myeloproliferative neoplasms: Disease sustainer and disease accelerator. Blood 2015, 125, 304–315. [Google Scholar] [CrossRef] [PubMed]
  56. Phase II Randomized Clinical Trial Comparing Ropeginterferon Versus Phlebotomy in Low-Risk Patients with Polycythemia Vera. Results of the Pre-Planned Interim Analysis. Available online: https://library.ehaweb.org/eha/2020/eha25th/303391/tiziano.barbui.phase.ii.randomized.clinical.trial.comparing.ropeginterferon.html?f=listing%3D0%2Abrowseby%3D8%2Asortby%3D1%2Asearch%3Dlb2602 (accessed on 14 June 2020).
  57. Gisslinger, H.; Buxhofer-Ausch, V.; Thaler, J.; Forjan, E.; Willenbacher, E.; Wolf, D.; Kralovics, R.; Gisslinger, B.; Stampfl-Mattersberger, M.; Egle, A.; et al. Long-Term Efficacy and Safety of Ropeginterferon Alfa-2b in Patients with Polycythemia Vera — Final Phase I/II Peginvera Study Results. Blood 2018, 132, 3030. [Google Scholar] [CrossRef]
  58. Silver, R.T.; Vandris, K.; Goldman, J.J. Recombinant interferon-α may retard progression of early primary myelofibrosis: A preliminary report. Blood 2011, 117, 6669–6672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Rank, C.U.; Bjerrum, O.W.; Larsen, T.S.; Kjær, L.; De Stricker, K.; Riley, C.H.; Hasselbalch, H.C. Minimal Residual Disease after Long-term Interferon-Alpha2 treatment. A Report on Hematological, Molecular, and Histomorphological Response Patterns in Ten Patients with Essential Thrombocythemia and Polycythemia Vera. Leuk. Lymphoma 2015, 57, 1–26. [Google Scholar] [CrossRef]
  60. Ianotto, J.-C.; Chauveau, A.; Boyer-Perrard, F.; Gyan, E.; Laribi, K.; Cony-Makhoul, P.; Demory, J.-L.; De Renzis, B.; Dosquet, C.; Rey, J.; et al. Benefits and pitfalls of pegylated interferon-α2a therapy in patients with myeloproliferative neoplasm-associated myelofibrosis: A French Intergroup of Myeloproliferative neoplasms (FIM) study. Haematologica 2017, 103, 438–446. [Google Scholar] [CrossRef] [Green Version]
  61. Silver, R.T.; Barel, A.C.; Lascu, E.; Ritchie, E.K.; Roboz, G.J.; Christos, P.J.; Orazi, A.; Hassane, D.; Tam, W.; Cross, N.C.P. The effect of initial molecular profile on response to recombinant interferon-α (rIFNα) treatment in early myelofibrosis. Cancer 2017, 123, 2680–2687. [Google Scholar] [CrossRef]
  62. Barosi, G.; Mesa, R.; Finazzi, G.; Harrison, C.; Kiladjian, J.-J.; Lengfelder, E.; McMullin, M.F.; Passamonti, F.; Vannucchi, A.M.; Besses, C.; et al. Revised response criteria for polycythemia vera and essential thrombocythemia: An ELN and IWG-MRT consensus project. Blood 2013, 121, 4778–4781. [Google Scholar] [CrossRef]
  63. Bjørn, M.E.; Hasselbalch, H.C. Minimal residual disease or cure in MPNs? Rationales and perspectives on combination therapy with interferon-alpha2 and ruxolitinib. Expert Rev. Hematol. 2017, 94, 1–12. [Google Scholar] [CrossRef] [PubMed]
  64. Sørensen, A.L.; Mikkelsen, S.U.; Knudsen, T.A.; Bjørn, M.E.; Andersen, C.L.; Bjerrum, O.W.; Brochmann, N.; Patel, D.A.; Gjerdrum, L.M.R.; El Fassi, D.; et al. Ruxolitinib and interferon-α2 combination therapy for patients with polycythemia vera or myelofibrosis: A phase II study. Haematologica 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kiladjian, J.-J.; Soret-Dulphy, J.; Resche-Rigon, M.; Boyer-Perrard, F.; Barraco, F.; Rolland-Neyret, V.; Capron, C.; Chevret, S.; Giraudier, S.; Cassinat, B. Ruxopeg, a Multi-Center Bayesian Phase 1/2 Adaptive Randomized Trial of the Combination of Ruxolitinib and Pegylated Interferon Alpha 2a in Patients with Myeloproliferative Neoplasm (MPN)-Associated Myelofibrosis. Blood 2018, 132, 581. [Google Scholar] [CrossRef]
  66. Barbui, T.; Ghirardi, A.; Masciulli, A.; Carobbio, A.; Palandri, F.; Vianelli, N.; De Stefano, V.; Betti, S.; Di Veroli, A.; Iurlo, A.; et al. Second cancer in Philadelphia negative myeloproliferative neoplasms (MPN-K). A nested case-control study. Leukemia 2019, 33, 1996–2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Mondello, P.; Di Mirto, C.; Cuzzocrea, S.; Arrigo, C.; Mian, M.; Pitini, V.; M, V.P. Interferon Alpha Has a Strong Anti-tumor Effect in Philadelphia-negative Myeloproliferative Neoplasms. Clin. Lymphoma Myeloma Leuk. 2019, 19, e489–e495. [Google Scholar] [CrossRef] [PubMed]
  68. Kiladjian, J.-J.; Chevret, S.; Dosquet, C.; Chomienne, C.; Rain, J.-D. Treatment of Polycythemia Vera With Hydroxyurea and Pipobroman: Final Results of a Randomized Trial Initiated in 1980. J. Clin. Oncol. 2011, 29, 3907–3913. [Google Scholar] [CrossRef]
  69. Srour, S.A.; Devesa, S.S.; Morton, L.M.; Check, D.P.; Curtis, R.E.; Linet, M.S.; Dores, G.M. Incidence and patient survival of myeloproliferative neoplasms and myelodysplastic/myeloproliferative neoplasms in the United States, 2001-12. Br. J. Haematol. 2016, 174, 382–396. [Google Scholar] [CrossRef] [Green Version]
  70. Griesshammer, M.; Sadjadian, P.; Wille, K. Contemporary management of patients withBCR-ABL1-negative myeloproliferative neoplasms during pregnancy. Expert Rev. Hematol. 2018, 11, 697–706. [Google Scholar] [CrossRef]
  71. How, C.-J.; Leiva, O.; Bogue, T.; Fell, G.; Bustoros, M.; Connell, N.T.; Connors, J.M.; Ghobrial, I.M.; Higgins, N.; Jessop, N.A.; et al. Pregnancy Outcomes, Risk Factors, and Gestational Cell Count Trends in Pregnant Women with Essential Thrombocythemia and Polycythemia Vera. Blood 2019, 134, 4172. [Google Scholar] [CrossRef]
  72. Skeith, L.; Carrier, M.; Robinson, S.; Alimam, S.; Rodger, M. Risk of venous thromboembolism in pregnant women with essential thrombocythemia: A systematic review and meta-analysis. Blood 2017, 129, 934–939. [Google Scholar] [CrossRef]
  73. Maze, D.; Kazi, S.; Gupta, V.; Malinowski, A.K.; Fazelzad, R.; Shah, P.S.; Shehata, N. Association of Treatments for Myeloproliferative Neoplasms During Pregnancy With Birth Rates and Maternal Outcomes. JAMA Netw. Open 2019, 2, e1912666. [Google Scholar] [CrossRef] [PubMed]
  74. Tefferi, A.; Passamonti, F. Essential thrombocythemia and pregnancy: Observations from recent studies and management recommendations. Am. J. Hematol. 2009, 84, 629–630. [Google Scholar] [CrossRef]
  75. Balsat, M.; Etienne, M.; Elhamri, M.; Hayette, S.; Salles, G.; Thomas, X. Successful pregnancies in patients with BCR-ABL-positive leukemias treated with interferon-alpha therapy during the tyrosine kinase inhibitors era. Eur. J. Haematol. 2018, 101, 774–780. [Google Scholar] [CrossRef]
  76. Pons, J.-C.; Lebon, P.; Frydman, R.; Delfraissy, J.-F. Pharmacokinetics of Interferon-Alpha in Pregnant Women and Fetoplacental Passage. Fetal Diagn. Ther. 1995, 10, 7–10. [Google Scholar] [CrossRef] [PubMed]
  77. Beauverd, Y.; Radia, D.; Cargo, C.; Knapper, S.; Drummond, M.; Pillai, A.; Harrison, C.; Robinson, S. Pegylated interferon alpha-2a for essential thrombocythemia during pregnancy: Outcome and safety. A case series. Haematologica 2016, 101, e182–e184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Kumar, A.R.; Hale, T.W.; Mock, R.E. Transfer of Interferon Alfa Into Human Breast Milk. J. Hum. Lact. 2000, 16, 226–228. [Google Scholar] [CrossRef]
Table 1. FDA Approvals and Recommendations for Use of Standard Interferon, Pegylated Interferon, and Ropeginterferon in MPN.
Table 1. FDA Approvals and Recommendations for Use of Standard Interferon, Pegylated Interferon, and Ropeginterferon in MPN.
FDA Approvals
Hairy cell leukemia
Adjuvant treatment for high-risk melanoma 1
Clinically aggressive follicular lymphoma, with anthracycline-containing chemotherapy 1
AIDS-related Kaposi sarcoma
Chronic hepatitis B
Condylomata acumunata
Consensus Opinions on Use in MPN
Low-risk myelofibrosis
Consider if symptomatic
High-risk polycythemia vera and essential thrombocythemia:
First-line treatment (as alternative to hydroxyurea) 2
Second-line treatment (if refractory to or intolerant of hydroxyurea) 2
Low-risk polycythemia vera and essential thrombocythemia
Consider if symptomatic and need for cytoreduction
Pregnant or breastfeeding MPN patients in need of cytoreduction
1 No longer clinically used. 2 National Comprehensive Cancer Network (NCCN) recommends consideration as first-line in young patients or patients considering pregnancy; European Leukemia Net (ELN) recommends interferon as acceptable first-line in high-risk polycythemia vera in need for cytoreduction.
Table 2. Summary of Clinical Trials Evaluating Single-Agent Pegylated Interferon Alfa 2A.
Table 2. Summary of Clinical Trials Evaluating Single-Agent Pegylated Interferon Alfa 2A.
Clinical TrialPhasePatientsComparator ArmClinical Response (RR/CR/PR)Molecular Response (RR/CR/PR)Discontinuation Rate
pIFNComparatorpIFNHUpIFNComparator
Masarova et al. 2017 [43]2ET (n = 40) and PV (n = 43)None80% A/75%/5%63%/18%/36%22%
DALIAH [49]3Untreated MPN (n = 205)Hydroxyurea49%/9%/33% A,B75%/19%/56% A,B17%/16%/1%NR/23%/NR27%5%
MPD-RC 112 [48,50]3High-risk ET (n = 39) and PV (n = 43)Hydroxyurea78%/35%/43% C70%/37%/32% C11% D/NR/NR20% D/NR/NR15%10%
MPN-RC 111 [47,48]2High-risk ET (n = 65) and PV (n = 50) resistant/refractory to hydroxyureaNone65%/34%/31% CNR14%
A Hematologic response; B ET, PV, and pre-MF patients only; C ELN response criteria; D cytogenetic response. CR: complete response; ET: essential thrombocythemia; MPN: myeloproliferative neoplasm; NR: not reported; PR: partial response; PV: polycythemia vera; pIFN: pegylated interferon; RR: response rate.
Table 3. Summary of Clinical trials Evaluating Ropeginterferon Alfa 2B.
Table 3. Summary of Clinical trials Evaluating Ropeginterferon Alfa 2B.
Clinical TrialPhasePatientsComparator ArmClinical Response (RR/CR/PR)Molecular Response (RR/CR/PR)Discontinuation Rate
rpIFNComparatorrpIFNComparatorrpIFNComparator
PEGINVERA [57]1/2PV (n = 51)None99%/64%/33% A74%/29%/45%41%
PROUD-PV [53]3PV with less than 3 years of cytoreduction (n = 257)Hydroxyurea21 B/43% A/NR28% B/46% A/NR34%/NR/NR42%/NR/NRNR
CONTINUATION-PV [53] 3PV with less than 3 years of cytoreduction (n = 257)Hydroxyurea53% C/71% A/NR38%/51% A/NR66%/NR/NR27%/NR/NR8%4%
LOW-PV [56]2Low-risk PV (n = 100)Phlebotomy84% D/NR/NR66%/NR/NRNRNRNR
A Hematologic response; B Composite hematologic response and spleen size reduction; C Complete hematologic response and improvement in disease burden at 3 years; D Response Criteria: hematocrit < 45% with no progressive disease. CR: complete response; NR: not reported; PR: partial response; PV: polycythemia vera; rpIFN: ropegylated interferon; RR: response rate.
Table 4. Summary of Clinical Trials Evaluating Combination Pegylated Interferon Alfa 2A and Ruxolitinib.
Table 4. Summary of Clinical Trials Evaluating Combination Pegylated Interferon Alfa 2A and Ruxolitinib.
Clinical TrialPhasePatientsComparator ArmClinical Response (RR/CR/PR)Molecular Response (RR/CR/PR)Discontinuation Rate
RUXOPEG [65]1/2Intermediate/high-risk MF (n = 15)None0%/NR/NR A30%/NR/NR ANRNR
COMBI [64]2PV (n = 32) and MF (n = 18) with active diseaseNone31%/9%/22% (PV) B
44%/28%/72% (MF) A
41%/2%/35%6% (PV)32% (MF)
A IWG-MRT Clinical response criteria; B ELN response criteria. CR: complete response; MF: myelofibrosis; NR: not reported; PR: partial response; PV: polycythemia vera; pIFN: pegylated interferon; RR: response rate.

Share and Cite

MDPI and ACS Style

How, J.; Hobbs, G. Use of Interferon Alfa in the Treatment of Myeloproliferative Neoplasms: Perspectives and Review of the Literature. Cancers 2020, 12, 1954. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12071954

AMA Style

How J, Hobbs G. Use of Interferon Alfa in the Treatment of Myeloproliferative Neoplasms: Perspectives and Review of the Literature. Cancers. 2020; 12(7):1954. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12071954

Chicago/Turabian Style

How, Joan, and Gabriela Hobbs. 2020. "Use of Interferon Alfa in the Treatment of Myeloproliferative Neoplasms: Perspectives and Review of the Literature" Cancers 12, no. 7: 1954. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers12071954

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop