Open access peer-reviewed chapter

Pharmacological Management of Idiopathic Pulmonary Fibrosis

Written By

Ladan Panahi, George Udeani, Andrew Scott Tenpas, Theresa Ofili, Elizabeth Marie Aguilar, Sarah Burchard, Alexandra Ruth Ritenour, April Jacob Chennat, Nehal Ahmed, Chairat Atphaisit, Crystal Chi, Jesus Cruz III, Monica D. Deleon, Samantha Lee, Zack Mayo, Mackenzie Mcbeth, Mariel Morales, Jennifer N. Nwosu, Kelly Palacios, Jaycob M. Pena and Nitza Vara

Submitted: 21 October 2021 Reviewed: 25 October 2021 Published: 17 December 2021

DOI: 10.5772/intechopen.101390

From the Edited Volume

Idiopathic Pulmonary Fibrosis

Edited by Salim Surani and Venkat Rajasurya

Chapter metrics overview

343 Chapter Downloads

View Full Metrics

Abstract

Idiopathic pulmonary fibrosis (IPF) is a common interstitial lung disease (ILD) caused by environmental exposures, infections, or traumatic injuries and subsequent epithelial damage. Since IPF is a progressively fatal disease without remission, treatment is both urgent and necessary. The two medications indicated solely for treatment include the tyrosine kinase inhibitor nintedanib (Ofev®) and the anti-fibrotic agent pirfenidone (Esbriet®). This chapter discusses in detail the current treatment options for clinical management of IPF, specifically the mentioned two pharmacotherapeutic agents that decrease physiological progression and likely improve progression-free survival. The chapter also discusses the evolution of drug therapy in IPF management and the drawbacks and limitations learned throughout historical trials and observational studies.

Keywords

  • drug therapy
  • pharmacological management
  • idiopathic pulmonary fibrosis
  • review

1. Introduction

Idiopathic pulmonary fibrosis (IPF) is a common interstitial lung disease (ILD) caused by environmental exposures, infections, or traumatic injuries and subsequent epithelial damage [1, 2]. It is characterized by fibroblast activation, followed by excessive secretion of extracellular matrix in the bronchial walls and alveolar interstitium [3]. This uncontrolled deposition leads to stiffening of lung tissue, which impairs diffusion of gases and reduces blood oxygenation [3, 4]. More prevalent among males and adults over 65 years old, it has a high incidence in North America and Europe [1]. Smoking, family history, and genetic mutations associated with telomere length maintenance have been linked to increased risk of developing IPF, as well as the history of gastroesophageal reflux disease and obstructive sleep apnea [1].

Patients typically present with chronic, progressive dyspnea, and dry cough [5]. Their history may include long-term smoke or workplace exposure such as inhalation of wood or metal particulates [6]. On physical examination, bibasilar inspiratory crackles (“velcro rales”) and finger clubbing may be seen [4, 7]. Pulmonary function tests (PFTs) usually demonstrate reduced lung capacity and reduced diffusion capacity for carbon monoxide, indicating restrictive disease and abnormal gas exchange [4, 8]. Exclusion of other interstitial lung diseases—including autoimmune diseases—is required before a diagnosis can be made. Additionally, the presence of a honeycomb fibrosis pattern on high-resolution computed tomography is necessary [4, 8]. Patients commonly have at least one comorbidity, such as chronic obstructive pulmonary disease (COPD), pulmonary hypertension, lung cancer, and diabetes mellitus [4, 8].

IPF is characterized by irreversible and potentially fatal lung deterioration [8, 9]. Patients may experience different rates of disease progression, ranging from gradual deterioration to stable periods lasting months or years. Symptoms associated with progression include worsening dyspnea, hypoxemia, and pulmonary hypertension, as well as fatal exacerbations, where respiratory function declines acutely and unpredictably [8, 9]. Although the disease course varies among patients, prognosis remain poor, with an average life expectancy of 3–5 years after diagnosis [4, 9].

Advertisement

2. Standards of care

Since IPF is a progressively fatal disease without remission, treatment is both urgent and necessary [10, 11]. The two medications indicated solely for treatment include the tyrosine kinase inhibitor nintedanib (Ofev®) and the anti-fibrotic agent pirfenidone (Esbriet®) [4, 12]. Both were approved in 2014 after clinical trials suggested that they halted the decline in lung function, including a decline in forced vital capacity (FVC) by 50% over a 1-year period [4, 11]. Moreover, they have been shown to be safe and effective in reducing severe respiratory episodes often seen in IPF [4].

Treatment regimens for COPD, heart disease, and smoking cessation are also recommended to reduce respiratory strain if experienced concurrently [4, 13]. Patients suffering from hypoxemia and IPF often receive supplemental oxygen [4, 14]. Pulmonary rehabilitation, physical therapy, and oxygen are all recommended to improve exercise tolerance and duration, reduce dyspnea, prevent the development of pulmonary hypertension, and improve overall lung capacity [4].

Lung transplantation remains a viable option for those who meet the criteria for the procedure [11]. It must be considered earlier in disease progression, with early evaluation to maximize eligibility [4, 11]. Past treatments like warfarin, N-acetylcysteine, prednisone, and azathioprine are no longer recommended due to an overall lack of treatment efficacy [4, 15]. Furthermore, these pharmacotherapeutic options should be avoided in IPF until high-quality randomized control trials prove efficacy since they have failed to show relevant reductive changes in FVC, adverse events, or death [16].

Advertisement

3. Non-pharmacological management and supportive care

Though current drug therapies demonstrate a reduction in acute exacerbations due to their cytotoxic and immunosuppressive side effect profiles, non-drug measures are often considered. Unfortunately, patients opting for mechanical ventilation—often as a bridge to lung transplantation—suffer from low survival rates [17]. Poor prognostic indicators include a decline in 6-minute walk (6 MW) distance greater than 150 meters within one year, a decrease in FVC greater than 10% within 6 months, and a decline in diffusing capacity for carbon monoxide (DLCO) greater than 15% within 6 months [4].

Improvements in both quality of life and 6 MW distance can be seen in those undergoing pulmonary rehabilitation [4]. Length of survival is highly variable; patients diagnosed with mild, moderate, and severe diseases survive an average of 55.6, 38.7, and 27.4 months, respectively [4]. Ultimately, transplantation remains the only option for those with advanced IPF; those who do not undergo this procedure often have poorer outcomes [4, 12]. Approximately 66% of transplant recipients live for more than 3 years postsurgery, while 53% survive greater than 5 years [4]. Transplantation does carry certain complications such as cancer, infections, primary graft dysfunction, cytomegalovirus, and allograft rejection are all commonly seen [12]. Moreover, supplemental oxygen has been shown to improve symptom control during exercise, while lung transplantation may increase survival rates and improve patients’ overall quality of life [14].

Since drug therapy is merely supportive therapy, patients are encouraged to take alternative measures to decrease their risk, including smoking cessation, supplemental oxygen, and pulmonary rehabilitation [4]. Ongoing GERD has been thought to worsen IPF, but the use of antacids based on clinical trials remains inconclusive [4]. Although the relation of GERD to IPF is still unknown, the prevalence of GERD and erosive esophagitis are observed more commonly in patients with IPF than in the general population [18, 19].

Lastly, patients should receive pneumonia and influenza vaccinations as part of complementary therapy, though there is no proven benefit for the previously mentioned interventions [14]. Although there is no documented outcome benefit with vaccination in the IPF setting, preventing pulmonary infections is essential as extrapulmonary comorbidities through interactions with environmental factors by various mechanisms are thought to contribute to IPF [20]. Vaccinations are especially recommended for post-transplantation patients since they may be more susceptible immunologically. In outpatient settings, pulmonary hypertension should be controlled with supplemental oxygen [21]. Unless a patient participates in a clinical trial, alternative therapies should be avoided.

Advertisement

4. Previous therapies

Although commonly used for their anti-inflammatory effects, corticosteroids do not improve clinical outcomes in IPF [14, 22]. When used as monotherapy, they show no survival benefit and actually increased risk of morbidity with long-term use [7, 14]. A regimen consisting of prednisone, azathioprine, and N-acetylcysteine (NAC) was once accepted therapy [7, 14]. However, trial data revealed that, compared to placebo, the combination increased risk of death and hospitalization [7, 14].

Ambrisentan (Letairis®), a potent type-A selective endothelin receptor antagonist, was once thought to decrease time to disease progression [7]. However, the ARTEMIS-IPF trial examined its use in IPF patients, finding it to be ineffective and associated with increased risk of hospitalizations and disease progression [7]. The trial was eventually terminated when an interim analysis found minimal efficacy [7]. Recent guidelines no longer recommend the anticoagulant warfarin since it was associated with a higher risk of mortality compared to placebo [7, 14].

Advertisement

5. Current therapy: nintedanib

After numerous studies yielded conflicting results, new treatment options were developed, including two novel anti-fibrotic agents capable of slowing disease progression [4]. Pirfenidone and nintedanib both demonstrated a significant reduction in annual FVC decline and improved survival [7].

Nintedanib (Ofev®), an oral tyrosine kinase inhibitor, inhibits the fibroblast proliferation leading to progression of lung fibrosis [3, 4]. It may also inhibit other growth factor receptors, including tyrosine kinase vascular endothelial growth factor receptor and platelet-derived growth factor receptor [3, 4, 7]. This multi-faceted inhibition makes it a first-line agent for IPF [3, 7]. The standard dose is 150 mg twice daily taken with food to increase bioavailability [4, 7]. However, dosing can be withheld or lowered to 100 mg twice daily if side effects become intolerable [7]. Once controlled, standard dosing can be resumed [7]. If adverse reactions persist, however, discontinuation should be considered [7]. The most common side effects associated with its use include diarrhea, nausea, and vomiting [7]. Other important side effects include weight loss and drug-induced hepatotoxicity, designated by a 3–5 fold increase in AST/ALT, with or without severe liver damage. Discontinuation or dose reduction is based on the presence of severe liver damage; details relating to specific therapeutic steps can be found in Table 1. Adverse reactions should be monitored alongside signs of increased bleeding, especially in those taking anticoagulants. Arterial thromboembolic events have been noted in patients taking nintedanib, and caution should be exercised in those at high risk for cardiovascular events [4]. Basic pharmacokinetics and special population dosing can be found in Table 1 [4].

Bioavailability5%
Increases by 20% when given with food
Half-life9.5 hours
Protein Binding97.8%
Volume of DistributionGreater than 1000 L
MetabolismHydrolytic cleavage by esterases (Major)
CYP3A4 (Minor)
EliminationMore than 90% of the dose eliminated via biliary/fecal excretion
Drug InteractionsP-glycoprotein (P-gp), CYP3A4 inducers
Dose AdjustmentsBaseline hepatic impairment:
  • Child-Pugh Class A: Reduce dose to 100 mg twice daily.

  • Child-Pugh Class B or C: Nintedanib not recommended.

Treatment-induced hepatotoxicity:
  • If AST or ALT increases to 3–5 times ULN, without signs of severe liver damage: Hold therapy or reduce dose to 100 mg twice daily. If values return to baseline, treatment may be restarted at a lower dose (100 mg twice daily), then increased to the full dose (150 mg twice daily).

  • If AST / ALT greater than three times ULN—with signs/symptoms of severe liver damage—or AST/ALT greater than five times ULN: Discontinue therapy

Monitoring Parameters
  • LFTs for the first 3 months of treatment

  • GI effects for first 3 months of treatment

  • Bleeding events

  • Cardiovascular events

  • Pregnancy test before initiation for those of childbearing age

Table 1.

Nintedanib pharmacokinetic parameters and special considerations [4].

Advertisement

6. Current therapy: pirfenidone

Pirfenidone (Esbriet®) is an oral synthetic pyridine derivative with anti-fibrotic and anti-inflammatory properties [7, 12, 23, 24]. Its anti-fibrotic effects arise from down-regulation of transforming growth factor (TGF) β and tumor necrosis factor (TNF) α [7, 23, 25]. It may inhibit fibroblast proliferation, expression of heat-shock protein 47, and collagen synthesis as well [7, 23, 24, 25]. Clinically, pirfenidone reduces worsening of FVC and may reduce risk of hospitalization [7, 23, 26]. Several studies like CAPACITY, ASCEND and RECAP have confirmed its long-term safety, efficacy, and favorable tolerability [7, 12].

Common side effects and clinical pharmacology can be found in Table 2. Most prevalent are gastrointestinal (GI) and skin-related adverse drug effects, which generally wane after the first 6 months and do not impact a patient’s ability to continue and maintain a high-dose intensity [12]. Several side effects like fatigue, photosensitivity, and GI distress may require dose reductions [7, 12]. Fatigue, in particular, is observed within the first few weeks of treatment and may substantially affect the quality of life. It may be difficult to distinguish from the disease itself, though it can be managed by dose modifications or even discontinuation [7, 12]. Several studies have examined the importance of taking pirfenidone with food [27, 28, 30, 31]. Administration after meals slows absorption and may mitigate GI side effects [7, 12, 27, 28].

BioavailabilityUnknown
Half-Life3.0 hours
Protein bindingMean of 50–58% at concentrations of 1–10 μg/mL.
Volume of DistributionMean of 59–71 L following oral administration
Dosage and AdministrationRecommend titration to 801 mg three times daily (2403 mg/day) with food
Metabolism and ExcretionPrimarily metabolized in the liver and bio-transformed by CYP1A2
Roughly 80% dose excreted in urine as metabolite 5-carboxy-pirfenidone
Common side effectsNausea, rash, dyspnea, diarrhea, fatigue, bronchitis, upper respiratory tract infections, dizziness, photosensitivity
Interactions
  • CYP1A2 inhibitors (ciprofloxacin, fluvoxamine) may decrease metabolism and require dosing adjustments or discontinuation.

  • Grapefruit juice should be used with caution, though study results are inconsistent.

Warnings/Precautions
  • Photosensitivity reactions may require dose adjustments.

  • Limit exposure to sunlight and sunlamps, use sunscreen (SPF ≥ 50) and protective clothing while taking.

  • GI side effects may be managed with temporary dose reduction, with gradual titration back to full dose. Taking after a full meal may help.

  • Mild-to-severe fatigue can be managed by dose modifications but may necessitate discontinuation.

  • Elevated liver enzymes (AST, ALT, bilirubin) occurred in trials and may require dose adjustments or discontinuation.

Monitoring Parameters
  • Monitor liver function (AST, ALT, bilirubin) before initiating and each month after for six months, then every three months, or if the patient experiences symptoms of liver injury.

  • If ALT or AST exceeds 3–5 times ULN—with no symptoms—dose adjustments may be made. If 3–5 times ULN—accompanied with symptoms or hyperbilirubinemia—or > 5× ULN, discontinue permanently.

Special Populations
  • Hepatic impairment: Monitor liver function monitored closely and potential for adverse reactions. Use contraindicated in those with severe hepatic dysfunction or end-stage liver disease

  • Renal impairment: Avoid severe kidney impairment (CrCl <30 mL/min) or dialysis.

  • Pregnant and nursing women: Not studied. It should be avoided during pregnancy and when nursing.

  • Geriatrics: No dose adjustments needed.

  • Pediatrics: Not studied.

Table 2.

Pharmacokinetic parameters and special considerations [7, 12, 27, 28, 29].

Updated practice guidelines recommend both nintedanib and pirfenidone [15]. Though both have been shown safe and effective, a lack of head-to-head trials makes it difficult to recommend one over the other [15]. The two agents have a different mechanism of action, making the prospect of combination therapy intriguing [12, 32]. However, when investigated, it was found that the combination led to greater photosensitivity and GI side effects [12, 32].

Advertisement

7. Acute exacerbations

Acute exacerbations (AE) are defined as an acute downturn in blood oxygenation, increased lung attenuation per computed tomography scan, and acute worsening of dyspnea [33]. Common causes include exposure to particulate matter (PM) ≥ 2.5 μm or crocin peptide released by S. nepalensis, bronchoscopy or lung biopsy, and inhalation of water repellant [33, 34, 35, 36]. Sources of PM include tobacco smoke, candles, forest fires, and dust [33, 37]. The exact incidence of exacerbations is unknown but is estimated to vary between 5 and 20% [36, 38].

Since AE mortality rates range between 60 and 80% within a 90-day period, most care is strictly palliative in nature [39]. The two primary therapies include corticosteroids like prednisone and cytotoxic medications like cyclophosphamide. However, no proven benefit for these therapies has been demonstrated [40]. In addition, mechanical ventilation should not be employed due to poor outcomes [41]. Novel therapy involving administration of polymyxin B-immobilized fiber column (PMX-DHP), originally developed to manage sepsis by removing plasma endotoxins, has shown increased effectiveness [38, 42]. One limitation of its use is it can lower white blood cell counts via absorption of neutrophils [38, 39]. It remains most effective if administered within 3–7 days of AE onset [38, 39].

Advertisement

8. Clinical evidence for efficacy

The SENSCIS trial was a 52-week randomized, placebo-controlled, double-blind study examining the treatment of systemic sclerosis-associated interstitial lung disease (SSc-ILD) with nintedanib [43, 44]. It was shown to decrease FVC decline rate (mL/year) within the treatment group compared to placebo [3, 43]. An annual difference of −52.4 mL/year for nintedanib versus −93.3 mL/year for placebo was shown at 52 weeks [3, 43]. The INPULSIS trial, a 52-week, randomized, double-blind, phase 3 trial, showed a similar reduction in FVC decline rate with nintedanib versus placebo [3, 45]. TOMORROW, a 52-week, randomized, double-blind, placebo-controlled, phase 2 trial—alongside INPULSIS—showed a decrease in acute exacerbations with nintedanib compared to placebo [4, 45]. The INBUILD trial, a 52-week, randomized, double-blind, placebo-controlled, parallel-group trial, examined patients with progressive fibrosing interstitial lung diseases other than IPF [3, 46]. Treatment groups received nintedanib 150 mg twice daily or placebo [3, 46]. Reduction in FVC decline rate was uniform across the five subgroups [46, 47].

Three major trials have recently examined pirfenidone, including CAPACITY 004, CAPACITY 006, and ASCEND [4, 12]. The two CAPACITY trials were run side-by-side for 72 weeks [48, 49]. CAPACITY 004 showed a significant reduction in FVC decline with pirfenidone, though only a significant difference up to week 48 was seen in CAPACITY 006 [48, 49]. The ASCEND trial, a 52-week, phase 3 trial, found that patients with a predicted FVC > 50% at baseline received benefit from pirfenidone over 1 year, reducing the rate of decline by approximately 50% [4, 49]. ASCEND also analyzed 6 MW distance [49, 50]. There was a significant difference between baseline and week 52 with the two treatments, including a 27.5% reduction in the pirfenidone group [49, 50]. Pooled population data from all three trials showed a 48% reduced risk of death at 1 year compared to placebo [48, 49].

Advertisement

9. Comparison: nintedanib vs. pirfenidone

As mentioned above, both agents have been shown safe and effective in placebo-controlled, randomized trials [51, 52, 53]. Both may slow the FVC decline rate by almost 50% over 1 year [51]. The two treatments have also demonstrated remarkable efficacy in minimizing severe respiratory hospitalizations and acute exacerbations [51, 54]. Though both agents may reduce mortality, each cost over $100,000 annually [51, 55].

9.1 Mechanisms of action

Nintedanib has a unique mechanism of action compared to pirfenidone. It inhibits tyrosine kinase, an enzyme that targets growth factor (GF) pathway receptors like fibroblast GF, platelet-derived GF, and vascular endothelial GF [3, 4]. Elevated bleeding risk is seen in patients taking concomitant anticoagulation therapy [7, 15]. Patients should regularly monitor liver function and GI disturbances, including diarrhea [43, 46]. Conversely, pirfenidone inhibits collagen synthesis, downregulates TGF-β, tumor necrosis factor-α, and reduces fibroblast proliferation [7, 24, 26]. Side effects include abnormal liver function, anorexia, nausea, photosensitive rashes, and vomiting [56].

9.2 Dosing

Pirfenidone comes in a 267 mg capsule, initially dosed as one capsule three times daily the first week [56, 57]. During the second week, the dose can be increased to 534 mg three times daily, and—after two weeks—it can be fully titrated to 801 mg three times (2,403 mg or nine pills per day). It is recommended that each dose be taken after a full meal to minimize GI side effects like nausea, dizziness, and vomiting [56, 57]. Patients may be treated with nintedanib first-line if intolerability to pirfenidone occurs [56, 57, 58]. The maximum recommended dose is 150 mg twice daily [58, 59].

9.3 Research similarities and differences

In a 1-year evaluation of both medications, there was a slight decrease in FVC, especially in those with comorbidities, which may account for increases in hospitalization and all-cause mortality [60, 61]. In combined studies, pirfenidone displayed a slower rate of FVC decline than nintedanib, helping to explain increased hospitalizations and mortality with its use [60, 61].

9.4 Side effect profiles

Though pirfenidone is frequently associated with GI complications, diarrhea, and involuntary weight loss is more common with nintedanib [57, 58, 61]. Pirfenidone’s side effects include dyspepsia, nausea, loss of appetite, phototoxic reactions, and difficulty concentrating. Sunscreen use is recommended when taking it. Conversely, nintedanib displays less nausea but greater transaminase elevations [57, 58, 61]. Pirfenidone is older and better studied more nintedanib, which may explain why its gastrointestinal and cognitive side effects are better understood [57, 58, 61]. Phototoxicity is generally absent with nintedanib [57, 61].

Advertisement

10. Therapeutic drawbacks

Though pirfenidone and nintedanib may slow disease progression, neither will cure IPF or markedly improve current symptoms [4, 62]. Symptom management, especially cough and dyspnea, is crucial to maintaining the quality of life [4, 63]. This is somewhat challenging given the lack of clinical evidence showing improvement in such symptoms and guideline focus on lung function [4, 63].

Both agents have noteworthy side effects. Nintedanib is most frequently associated with diarrhea, nausea, vomiting, and elevated liver enzymes [62]. By comparison, pirfenidone may cause nausea, diarrhea, dyspepsia, anorexia, and gastroesophageal reflux, as well as rash, upper respiratory infections, and fatigue [62, 64].

Another significant burden of IPF is cost [4, 65]. A recent systematic review estimated its annual cost in the United States at $20,000 per patient per year, about three times greater than the national health care resource use per capita [4, 65]. Hospitalizations and acute exacerbations are key drivers of this cost, with an average cost exceeding $16,000 for each IPF-related hospitalization [4, 66]. Due to their specialty drug and brand-only status, pirfenidone and nintedanib remain extremely expensive, with costs exceeding $10,000 per month per agent [4]. However, nintedanib is associated with fewer acute exacerbations and, consequently, decreased medical costs [4, 62, 67]. A recent comparison analysis from the United Kingdom found that the two drugs were comparable in estimated cost and health-related quality of life benefit [4, 68].

It is important to remember that the INBUILD trial was not powered to provide sufficient evidence for the use of nintedanib in rarer, specific fibrosing ILD [47]. However, it can be challenging to recruit patients with these rarer disease states. The fact that nintedanib reduced the rate of disease progression (i.e., FVC decline) in a wide range of progressive fibrosing ILD suggests utility in such populations [47].

11. Novel research/pipeline drugs

In recent decades, our understanding of IPF pathogenicity and management has improved significantly [15, 69]. However, many limitations, such as an inability to translate experimental findings in animal models to human subjects, remain a challenge [69, 70]. Current therapies like nintedanib and pirfenidone are limited to pathways involved in reducing disease progression and physiological decline in those with mild-to-moderate impairment [15, 69]. Second-line treatments capable of improving functional capacity for such patients or benefiting the severely impaired are still needed [15, 70].

Other viable agents have been recently investigated [9]. Increased concentrations of endothelin receptors have been observed in IPF lung tissue [15]. As a result, several clinically significant endothelin receptor antagonists have been previously tested, including ambrisentan, a selective type-A antagonist, and bosentan and macitentan, type-A and type-B antagonists [71, 72]. Nonetheless, recent guidelines strongly discourage the use of ambrisentan given its risk of harm and lack of benefit, along with a conditional recommendation against the use of bosentan and macitentan [15, 73].

The phosphodiesterase-5 inhibitor sildenafil has been investigated due to its role in pulmonary vasodilation and improved gas exchange [15]. Past studies and analyses reported a slight but significant improvement in the degree of dyspnea and quality of life compared to placebo [15]. However, it has failed to demonstrate improvements in mortality, acute exacerbations, and adverse events [15]. Recent guidelines discourage its use, though it continues to be investigated [15, 73].

N-acetylcysteine (NAC), a precursor of the antioxidant glutathione, has also been examined for use in IPF [74]. A pooled analysis compared NAC monotherapy to placebo in IPF patients [15]. Ultimately, there was no significant difference in the rate of death or acute exacerbation, as well as no significant benefit in mortality, quality of life, or adverse outcomes [15]. Current guidelines strongly discourage its use in practice [15, 74].

A recent randomized clinical trial investigated imatinib mesylate (Gleevec®), a tyrosine kinase inhibitor. It showed a statistically significant increased risk of adverse events and no improvement in preventing disease progression or mortality [15, 74]. This distinct lack of benefit has led to its use being discouraged in IPF [15, 74].

Several active interventional and observational trials are currently underway. Recent novel studies suggest that genetic factors may play a crucial role in overall risk, disease progression, and therapeutic response [70, 75]. Future trials and drug development will likely focus more on genetic variation in IPF patients [70, 75].

12. Conclusions

IPF is a common ILD that is progressive and potentially fatal [4, 9]. It is characterized by decreased lung function stemming from abnormal fibrotic processes, ultimately leading to scarring tissue formation, diminished gas exchange, and reduced blood oxygenation [4, 7]. Though there is no known cause, it is more common in males and elderly patients and is associated with risk factors like smoking, environmental exposure, and multiple comorbidities [4, 9]. Due to insufficient understanding of its pathophysiological mechanisms, there are currently no therapies capable of preventing or reversing IPF [7, 9, 76]. Current management includes antifibrotic drugs like nintedanib (Ofev®) and pirfenidone (Esbriet®), which have been shown to slow lung deterioration [4, 7, 13]. Recent investigations examining nintedanib use in other ILDs with progressive phenotypes have shown favorable results, suggesting that such ILDs share similar mechanisms and may thus benefit from similar treatment [3, 77]. Imatinib mesylate (Gleevec®) is not recommended due to the increased risk of adverse events and no improvement in disease progression or mortality [17, 73]. Similarly, the use of ambrisentan (Letairis®) is discouraged due to lack of effectiveness and increased risk of hospitalization [7]. Other IPF management strategies include smoking cessation, immunization, respiratory rehabilitation, oxygen supplementation, and management of comorbidities [4, 20]. More recent approaches have targeted biological processes linked to IPF, such as aging, oxidative stress, and epithelial-to-mesenchymal cell transition (EMT) [9, 76]. Ultimately, a better understanding of its underlying mechanisms is necessary to develop more effective treatments and reduce mortality [7, 9, 76].

References

  1. 1. Lederer DJ, Martinez FJ. Idiopathic pulmonary fibrosis. The New England Journal of Medicine. 2018;379(8):797-798. DOI: 10.1056/NEJMc1807508
  2. 2. Thannickal VJ, Toews GB, White ES, Lynch JP 3rd, Martinez FJ. Mechanisms of pulmonary fibrosis. Annual Review of Medicine. 2004;55:395-417. DOI: 10.1146/annurev.med.55.091902.103810
  3. 3. Wollin L, Distler JHW, Redente EF, Riches DWH, Stowasser S, Schlenker-Herceg R, et al. Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. European Respiratory Journal. 2019;54(3):1900161. DOI: 10.1183/13993003.00161-2019
  4. 4. Pleasants R, Tighe RM. Management of idiopathic pulmonary fibrosis. The Annals of Pharmacotherapy. 2019;53(12):1238-1248. DOI: 10.1177/1060028019862497
  5. 5. Van Manen MJ, Birring SS, Vancheri C, Cottin V, Renzoni EA, Russell A-M, et al. Cough in idiopathic pulmonary fibrosis. European Respiratory Review. 2016;25(141):278-286
  6. 6. Park Y, Ahn C, Kim T-HJS. Occupational and environmental risk factors of idiopathic pulmonary fibrosis: A systematic review and meta-analyses. Scientific Reports. 2021;11(1):1-10
  7. 7. Glass DS, Grossfeld D, Renna HA, Agarwala P, Spiegler P, Kasselman LJ, et al. Idiopathic pulmonary fibrosis: Molecular mechanisms and potential treatment approaches. Respiratory Investigation. 2020;58(5):320-335. DOI: 10.1016/j.resinv.2020.04.002
  8. 8. Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nature Reviews Disease Primers. 2017;3:17074. DOI: 10.1038/nrdp.2017.74
  9. 9. Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, et al. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cellular and Molecular Life Sciences. 2021;78(5):2031-2057. DOI: 10.1007/s00018-020-03693-7
  10. 10. Ley B, Collard HR, King TE Jr. Clinical course and prediction of survival in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2011;183(4):431-440. DOI: 10.1164/rccm.201006-0894CI
  11. 11. Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389(10082):1941-1952. DOI: 10.1016/S0140-6736(17)30866-8
  12. 12. Lancaster LH, de Andrade JA, Zibrak JD, Padilla ML, Albera C, Nathan SD, et al. Pirfenidone safety and adverse event management in idiopathic pulmonary fibrosis. European Respiratory Review. 2017;26(146):170057. DOI: 10.1183/16000617.0057-2017
  13. 13. Sgalla G, Iovene B, Calvello M, Ori M, Varone F, Richeldi L. Idiopathic pulmonary fibrosis: Pathogenesis and management. Respiratory Research. 2018;19(1):32. DOI: 10.1186/s12931-018-0730-2
  14. 14. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, et al. An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. American Journal of Respiratory and Critical Care Medicine. 2011;183(6):788-824. DOI: 10.1164/rccm.2009-040GL
  15. 15. Raghu G, Rochwerg B, Zhang Y, Cuello Garcia CA, Azuma A, Behr J, et al. An Official ATS/ERS/JRS/ALAT clinical practice guideline: Treatment of idiopathic pulmonary fibrosis. An update of the 2011 clinical practice guideline. American Journal of Respiratory and Critical Care Medicine. 2015;192(2):e3-e19. DOI: 10.1164/rccm.201506-1063ST
  16. 16. Sun T, Liu J. Efficacy of N-acetylcysteine in idiopathic pulmonary fibrosis: A systematic review and meta-analysis. Medicine. 2016;95(19):e3629
  17. 17. Mooney JJ, Raimundo K, Chang E, Broder MS. Mechanical ventilation in idiopathic pulmonary fibrosis: A nationwide analysis of ventilator use, outcomes, and resource burden. BMC Pulmonary Medicine. 2017;17(1):1-9
  18. 18. Raghu G, Freudenberger TD, Yang S, Curtis JR, Spada C, Hayes J, et al. High prevalence of abnormal acid gastro-oesophageal reflux in idiopathic pulmonary fibrosis. The European Respiratory Journal. 2006;27(1):136-142. DOI: 10.1183/09031936.06.00037005
  19. 19. el-Serag HB, Sonnenberg A. Comorbid occurrence of laryngeal or pulmonary disease with esophagitis in United States military veterans. Gastroenterology. 1997;113(3):755-760. DOI: 10.1016/s0016-5085(97)70168-9
  20. 20. Luppi F, Kalluri M, Faverio P, Kreuter M, Ferrara G. Idiopathic pulmonary fibrosis beyond the lung: Understanding disease mechanisms to improve diagnosis and management. Respiratory Research. 2021;22(1):109. DOI: 10.1186/s12931-021-01711-1
  21. 21. Acharya G, Arya S, Badal S, Kumar D, Samdariya S. Pharmacotherapy in idiopathic pulmonary fibrosis. International Journal of Basic & Clinical Pharmacology. 2014;3(5):761
  22. 22. Papiris SA, Kagouridis K, Kolilekas L, Papaioannou AI, Roussou A, Triantafillidou C, et al. Survival in Idiopathic pulmonary fibrosis acute exacerbations: The non-steroid approach. BMC Pulmonary Medicine. 2015;15:162. DOI: 10.1186/s12890-015-0146-4
  23. 23. Noble PW, Albera C, Bradford WZ, Costabel U, Glassberg MK, Kardatzke D, et al. Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): Two randomised trials. Lancet. 2011;377(9779):1760-1769. DOI: 10.1016/S0140-6736(11)60405-4
  24. 24. Shi S, Wu J, Chen H, Chen H, Wu J, Zeng F. Single- and multiple-dose pharmacokinetics of pirfenidone, an antifibrotic agent, in healthy Chinese volunteers. Journal of Clinical Pharmacology. 2007;47(10):1268-1276. DOI: 10.1177/0091270007304104
  25. 25. Cho ME, Kopp JB. Pirfenidone: An anti-fibrotic therapy for progressive kidney disease. Expert Opinion on Investigational Drugs. 2010;19(2):275-283. DOI: 10.1517/13543780903501539
  26. 26. Ley B, Swigris J, Day BM, Stauffer JL, Raimundo K, Chou W, et al. Pirfenidone reduces respiratory-related hospitalizations in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2017;196(6):756-761. DOI: 10.1164/rccm.201701-0091OC
  27. 27. Hu J, Shang D, Xu X, He X, Ni X, Zhang M, et al. Effect of grapefruit juice and food on the pharmacokinetics of pirfenidone in healthy Chinese volunteers: A diet-drug interaction study. Xenobiotica. 2016;46(6):516-521. DOI: 10.3109/00498254.2015.1089365
  28. 28. Rubino CM, Bhavnani SM, Ambrose PG, Forrest A, Loutit JS. Effect of food and antacids on the pharmacokinetics of pirfenidone in older healthy adults. Pulmonary Pharmacology & Therapeutics. 2009;22(4):279-285. DOI: 10.1016/j.pupt.2009.03.003
  29. 29. Genentech I. Esbriet (Pirfenidone) Capsules and Film-Coated Tablets, for Oral Use [Package Insert]. South San Francisco; 2017
  30. 30. Barranco-Garduno LM, Buendia-Roldan I, Rodriguez JJ, González-Ramírez R, Cervantes-Nevárez AN, Neri-Salvador JC, et al. Pharmacokinetic evaluation of two pirfenidone formulations in patients with idiopathic pulmonary fibrosis and chronic hypersensitivity pneumonitis. Heliyon. 2020;6(10):e05279. DOI: 10.1016/j.heliyon.2020.e05279
  31. 31. Pan L, Belloni P, Ding HT, Wang J, Rubino CM, Putnam WS. A pharmacokinetic bioequivalence study comparing pirfenidone tablet and capsule dosage forms in healthy adult volunteers. Advances in Therapy. 2017;34(9):2071-2082. DOI: 10.1007/s12325-017-0594-8
  32. 32. Ogura T, Taniguchi H, Azuma A, Inoue Y, Kondoh Y, Hasegawa Y, et al. Safety and pharmacokinetics of nintedanib and pirfenidone in idiopathic pulmonary fibrosis. The European Respiratory Journal. 2015;45(5):1382-1392. DOI: 10.1183/09031936.00198013
  33. 33. Tahara M, Fujino Y, Yamasaki K, Oda K, Kido T, Sakamoto N, et al. Exposure to PM2.5 is a risk factor for acute exacerbation of surgically diagnosed idiopathic pulmonary fibrosis: A case-control study. Respiratory Research. 2021;22(1):80. DOI: 10.1186/s12931-021-01671-6
  34. 34. Bennett D, Bargagli E, Refini RM, Pieroni M, Fossi A, Romeo R, et al. Acute exacerbation of idiopathic pulmonary fibrosis after inhalation of a water repellent. International Journal of Occupational Medicine and Environmental Health. 2015;28(4):775-779. DOI: 10.13075/ijomeh.1896.00462
  35. 35. D'Alessandro-Gabazza CN, Kobayashi T, Yasuma T, Toda M, Kim H, Fujimoto H, et al. A Staphylococcus pro-apoptotic peptide induces acute exacerbation of pulmonary fibrosis. Nature Communications. 2020;11(1):1539. DOI: 10.1038/s41467-020-15344-3
  36. 36. Hyzy R, Huang S, Myers J, Flaherty K, Martinez F. Acute exacerbation of idiopathic pulmonary fibrosis. Chest. 2007;132(5):1652-1658. DOI: 10.1378/chest.07-0299
  37. 37. Tahara M, Fujino Y, Yamasaki K, Oda K, Kido T, Sakamoto N, et al. Exposure to PM 2.5 is a risk factor for acute exacerbation of surgically diagnosed idiopathic pulmonary fibrosis: A case–control study. Respiratory Research. 2021;22(1):1-11
  38. 38. Kishaba T. Acute exacerbation of idiopathic pulmonary fibrosis. Medicina (Kaunas). 2019;55(3):70. DOI: 10.3390/medicina55030070
  39. 39. Kim SY, Park JH, Kim HJ, Jang HJ, Kim HK, Kim SH, et al. Direct hemoperfusion with polymyxin B-immobilized fiber column in a patient with acute exacerbation of idiopathic pulmonary fibrosis. Acute and Critical Care. 2020;35(4):302-306. DOI: 10.4266/acc.2020.00038
  40. 40. Innabi A, Gomez-Manjarres D, Alzghoul BN, Chizinga M, Mehrad B, Patel DC. Cyclophosphamide for the treatment of acute exacerbation of interstitial lung disease: A review of the literature. Sarcoidosis, Vasculitis, and Diffuse Lung Diseases. 2021;38(1):e2021002
  41. 41. Luo Z, Yang L, Liu S, Hu Y, Cao Z, Zhu J, et al. Mechanical ventilation for acute respiratory failure due to idiopathic pulmonary fibrosis versus connective tissue disease-associated interstitial lung disease: Effectiveness and risk factors for death. The Clinical Respiratory Journal. 2020;14(10):918-932
  42. 42. Enomoto N, Mikamo M, Oyama Y, Kono M, Hashimoto D, Fujisawa T, et al. Treatment of acute exacerbation of idiopathic pulmonary fibrosis with direct hemoperfusion using a polymyxin B-immobilized fiber column improves survival. BMC Pulmonary Medicine. 2015;15:15. DOI: 10.1186/s12890-015-0004-4
  43. 43. Distler O, Highland KB, Gahlemann M, Azuma A, Fischer A, Mayes MD, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. The New England Journal of Medicine. 2019;380(26):2518-2528. DOI: 10.1056/NEJMoa1903076
  44. 44. Maher TM, Mayes MD, Kreuter M, Volkmann ER, Aringer M, Castellvi I, et al. Effect of nintedanib on lung function in patients with systemic sclerosis-associated interstitial lung disease: Further analyses of a randomized, double-blind placebo-controlled trial. Arthritis & Rheumatology. 2021;73(4):671-676. DOI: 10.1002/art.41576
  45. 45. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. The New England Journal of Medicine. 2014;370(22):2071-2082. DOI: 10.1056/NEJMoa1402584
  46. 46. Wells AU, Flaherty KR, Brown KK, Inoue K, Devaraj A, Richeldi L, et al. Nintedanib in patients with progressive fibrosing interstitial lung diseases-subgroup analyses by interstitial lung disease diagnosis in the INBUILD trial: A randomised, double-blind, placebo-controlled, parallel-group trial. The Lancet Respiratory Medicine. 2020;8(5):453-460. DOI: 10.1016/S2213-2600(20)30036-9
  47. 47. Flaherty KR, Fell CD, Huggins JT, Nunes H, Sussman R, Valenzuela C, et al. Safety of nintedanib added to pirfenidone treatment for idiopathic pulmonary fibrosis. European Respiratory Journal. 2018;52(2). DOI: 10.1183/13993003.00230-2018
  48. 48. Gulati S, Luckhardt TR. Updated evaluation of the safety, efficacy and tolerability of pirfenidone in the treatment of idiopathic pulmonary fibrosis. Drug Healthcare and Patient Safety. 2020;12:85-94. DOI: 10.2147/DHPS.S224007
  49. 49. King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. The New England Journal of Medicine. 2014;370(22):2083-2092. DOI: 10.1056/NEJMoa1402582
  50. 50. Nathan SD, Costabel U, Albera C, Behr J, Wuyts WA, Kirchgaessler K-U, et al. Pirfenidone in patients with idiopathic pulmonary fibrosis and more advanced lung function impairment. Respiratory Medicine. 2019;153:44-51. DOI: 10.1016/j.rmed.2019.04.016
  51. 51. Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. Diagnosis of idiopathic pulmonary fibrosis. An official ATS/ERS/JRS/ALAT clinical practice guideline. American Journal of Respiratory and Critical Care Medicine. 2018;198(5):e44-e68
  52. 52. Varone F, Sgalla G, Iovene B, Bruni T, Richeldi L. Nintedanib for the treatment of idiopathic pulmonary fibrosis. Expert Opinion on Pharmacotherapy. 2018;19(2):167-175
  53. 53. Saito S, Alkhatib A, Kolls JK, Kondoh Y, Lasky JA. Pharmacotherapy and adjunctive treatment for idiopathic pulmonary fibrosis (IPF). Journal of Thoracic Disease. 2019;11(Suppl 14):S1740
  54. 54. Dempsey TM, Sangaralingham LR, Yao X, Sanghavi D, Shah ND, Limper AH. Clinical effectiveness of antifibrotic medications for idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2019;200(2):168-174
  55. 55. Owens GM. Strategies to manage costs in idiopathic pulmonary fibrosis. The American Journal of Managed Care. 2017;23(Suppl 11):S191-S196
  56. 56. Costabel U, Bendstrup E, Cottin V, Dewint P, Egan JJJ, Ferguson J, et al. Pirfenidone in idiopathic pulmonary fibrosis: Expert panel discussion on the management of drug-related adverse events. Advances in Therapy. 2014;31(4):375-391. DOI: 10.1007/s12325-014-0112-1
  57. 57. Meyer KC, Decker CA. Role of pirfenidone in the management of pulmonary fibrosis. Therapeutics and Clinical Risk Management. 2017;13:427-437. DOI: 10.2147/TCRM.S81141
  58. 58. Brunnemer E, Walscher J, Tenenbaum S, Hausmanns J, Schulze K, Seiter M, et al. Real-world experience with nintedanib in patients with idiopathic pulmonary fibrosis. Respiration. 2018;95(5):301-309. DOI: 10.1159/000485933
  59. 59. Fukihara J, Kondoh Y. Nintedanib (OFEV) in the treatment of idiopathic pulmonary fibrosis. Expert Review of Respiratory Medicine. 2016;10(12):1247-1254. DOI: 10.1080/17476348.2016.1249854
  60. 60. Belhassen M, Dalon F, Nolin M, Van Ganse E. Comparative outcomes in patients receiving pirfenidone or nintedanib for idiopathic pulmonary fibrosis. Respiratory Research. 2021;22(1):135. DOI: 10.1186/s12931-021-01714-y
  61. 61. Cerri S, Monari M, Guerrieri A, Donatelli P, Bassi I, Garuti M, et al. Real-life comparison of pirfenidone and nintedanib in patients with idiopathic pulmonary fibrosis: A 24-month assessment. Respiratory Medicine. 2019;159:105803. DOI: 10.1016/j.rmed.2019.105803
  62. 62. Tzouvelekis A, Bonella F, Spagnolo P. Update on therapeutic management of idiopathic pulmonary fibrosis. Therapeutics and Clinical Risk Management. 2015;11:359-370. DOI: 10.2147/TCRM.S69716
  63. 63. Ferrara G, Luppi F, Birring SS, Cerri S, Caminati A, Sköld M, et al. Best supportive care for idiopathic pulmonary fibrosis: Current gaps and future directions. European Respiratory Review. 2018;27(147). DOI: 10.1183/16000617.0076-2017
  64. 64. Rahaghi FF, Safdar Z, Brown AW, de Andrade JA, Flaherty KR, Kaner RJ, et al. Expert consensus on the management of adverse events and prescribing practices associated with the treatment of patients taking pirfenidone for idiopathic pulmonary fibrosis: A Delphi consensus study. BMC Pulmonary Medicine. 2020;20(1):191. DOI: 10.1186/s12890-020-01209-4
  65. 65. Diamantopoulos A, Wright E, Vlahopoulou K, Cornic L, Schoof N, Maher TM. The burden of illness of idiopathic pulmonary fibrosis: A comprehensive evidence review. PharmacoEconomics. 2018;36(7):779-807. DOI: 10.1007/s40273-018-0631-8
  66. 66. Mooney JJ, Raimundo K, Chang E, Broder MS. Hospital cost and length of stay in idiopathic pulmonary fibrosis. Journal of Medical Economics. 2017;20(5):518-524. DOI: 10.1080/13696998.2017.1282864
  67. 67. Bonella F, Stowasser S, Wollin L. Idiopathic pulmonary fibrosis: Current treatment options and critical appraisal of nintedanib. Drug Design, Development and Therapy. 2015;9:6407-6419. DOI: 10.2147/DDDT.S76648
  68. 68. Rinciog C, Watkins M, Chang S, Maher TM, LeReun C, Esser D, et al. A cost-effectiveness analysis of nintedanib in idiopathic pulmonary fibrosis in the UK. PharmacoEconomics. 2017;35(4):479-491. DOI: 10.1007/s40273-016-0480-2
  69. 69. Raghu G. Idiopathic pulmonary fibrosis: Lessons from clinical trials over the past 25 years. European Respiratory Journal. 2017;50(4):1701209. DOI: 10.1183/13993003.01209-2017
  70. 70. Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nature Reviews Drug Discovery. 2017;16(11):810. DOI: 10.1038/nrd.2017.225
  71. 71. King TE Jr, Brown KK, Raghu G, du Bois RM, Lynch DA, Martinez F, et al. BUILD-3: A randomized, controlled trial of bosentan in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2011;184(1):92-99. DOI: 10.1164/rccm.201011-1874OC
  72. 72. Raghu G, Million-Rousseau R, Morganti A, Perchenet L, Behr J, Music Study Group. Macitentan for the treatment of idiopathic pulmonary fibrosis: The randomised controlled MUSIC trial. The European Respiratory Journal. 2013;42(6):1622-1632. DOI: 10.1183/09031936.00104612
  73. 73. Behr J, Gunther A, Bonella F, Geissler K, Koschel D, Kreuter M, et al. German guideline for idiopathic pulmonary fibrosis - Update on Pharmacological Therapies 2017. S2k-Leitlinie Idiopathische Lungenfibrose - Update zur medikamentosen Therapie 2017. Pneumologie. 2018;72(2):155-168. DOI: 10.1055/s-0043-123035
  74. 74. Antoniou KM, Margaritopoulos GA, Siafakas NM. Pharmacological treatment of idiopathic pulmonary fibrosis: From the past to the future. European Respiratory Review. 2013;22(129):281-291. DOI: 10.1183/09059180.00002113
  75. 75. Kaur A, Mathai SK, Schwartz DA. Genetics in idiopathic pulmonary fibrosis pathogenesis, prognosis, and treatment. Frontiers in Medicine (Lausanne). 2017;4:154. DOI: 10.3389/fmed.2017.00154
  76. 76. Veith C, Boots AW, Idris M, van Schooten FJ, van der Vliet A. Redox imbalance in idiopathic pulmonary fibrosis: A role for oxidant cross-talk between NADPH oxidase enzymes and mitochondria. Antioxidants & Redox Signaling. 2019;31(14):1092-1115. DOI: 10.1089/ars.2019.7742
  77. 77. Valenzuela C, Torrisi SE, Kahn N, Quaresma M, Stowasser S, Kreuter M. Ongoing challenges in pulmonary fibrosis and insights from the nintedanib clinical programme. Respiratory Research. 2020;21(1):7. DOI: 10.1186/s12931-019-1269-6

Written By

Ladan Panahi, George Udeani, Andrew Scott Tenpas, Theresa Ofili, Elizabeth Marie Aguilar, Sarah Burchard, Alexandra Ruth Ritenour, April Jacob Chennat, Nehal Ahmed, Chairat Atphaisit, Crystal Chi, Jesus Cruz III, Monica D. Deleon, Samantha Lee, Zack Mayo, Mackenzie Mcbeth, Mariel Morales, Jennifer N. Nwosu, Kelly Palacios, Jaycob M. Pena and Nitza Vara

Submitted: 21 October 2021 Reviewed: 25 October 2021 Published: 17 December 2021